Language selection

Search

Patent 2834589 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2834589
(54) English Title: METHOD FOR PREPARING FC-CONTAINING POLYPEPTIDES HAVING IMPROVED PROPERTIES
(54) French Title: PROCEDE DE PREPARATION DE POLYPEPTIDES CONTENANT FC A PROPRIETES AMELIOREES
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C7K 16/00 (2006.01)
  • A61K 39/395 (2006.01)
  • A61P 29/00 (2006.01)
  • C7K 16/24 (2006.01)
  • C7K 16/42 (2006.01)
  • C7K 19/00 (2006.01)
  • C12P 21/00 (2006.01)
(72) Inventors :
  • ZHA, DONGXING (United States of America)
(73) Owners :
  • MERCK SHARP & DOHME CORP.
(71) Applicants :
  • MERCK SHARP & DOHME CORP. (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2012-05-22
(87) Open to Public Inspection: 2012-11-29
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2012/038915
(87) International Publication Number: US2012038915
(85) National Entry: 2013-10-28

(30) Application Priority Data:
Application No. Country/Territory Date
61/489,743 (United States of America) 2011-05-25

Abstracts

English Abstract

The present invention is directed to methods and compositions for the production of Fc-containing polypeptides comprising mutations at positions 243, 264, 267 and 328 of the Fc region.


French Abstract

L'invention concerne des procédés et des compositions pour produire des polypeptides contenant Fc présentant des mutations aux positions 243, 264, 267 et 328 de la région Fc.

Claims

Note: Claims are shown in the official language in which they were submitted.


WHAT IS CLAIMED:
1) An Fc-containing polypeptide comprising mutations at amino acid positions
243, 264,
267 and 328 of the Fc region, wherein the numbering is according to the EU
index as in
Kabat.
2) The Fc-containing polypeptide of claim 1, wherein the mutations are F243A,
V264A,
S267E, and L328F.
3) The Fc-containing polypeptide of claim 1, comprising the amino acid
sequence of SEQ ID
NO:7, SEQ ID NO:8, or SEQ ID NO:17.
4) The Fc-containing polypeptide of claim 1, wherein said Fc-containing
polypeptide is an
antibody or an antibody fragment comprising sialylated N-glycans, wherein the
sialic acid
residues in the sialylated N-glycans are attached via .alpha.-2,6 linkages.
5) The Fc-containing polypeptide of claim 1, wherein said Fc-containing
polypeptide is an
antibody or an antibody fragment comprising sialylated N-glycans comprising a
structure
selected from SA(1-4)Gal(1-4)GlcNAc(2-4)Man3GlcNAc2 or
SAGalGlcNAcMan5GlcNAQ , wherein the sialic acid residues are attached via
.alpha.-2,6
linkages.
6) The Fc-containing polypeptide of claim 1, wherein said Fc-containing
polypeptide has
one or more of the following properties when compared to a parent Fc-
containing
polypeptide:
a) reduced effector function;
b) increased anti-inflammatory properties;
c) increased binding to a lectin (e.g., CD22 (Siglec 2));
d) reduced binding to Fc.gamma.RIIa;
e) increased binding to Fc.gamma.RIIb;
f) reduced binding to Fc.gamma.RIIIa;
g) reduced binding to Fc.gamma.RIIIb.
7) A method for producing a Fc-containing polypeptide in a host cell
comprising:
-70-

a) providing a cell that has been genetically engineered to produce an Fc-
containing
polypeptide, wherein the host cell comprises a nucleic acid encoding mutations
at
amino acid positions 243, 264, 267 and 328 of the Fc region, wherein the
numbering
is according to the EU index as in Kabat;
b) culturing the host cell under conditions which cause expression of the Fc-
containing
polypeptide; and
c) isolating the Fe-containing polypeptide from the host cell.
8) The method of claim 7, wherein the nucleic acid encodes the mutations
F243A, V264A,
S267E, and L328F.
9) The method of claim 7, werein the Fc-containing polypeptide comprises the
amino acid
sequence of SEQ ID NO:7, SEQ ID NO:8, or SEQ ID NO:17.
10) The method of claim 7, wherein the Fc-containing polypeptide is an
antibody or an antibody
fragment comprising sialylated N-glycans, wherein tthe sialylated N-glycans
are attached via
.alpha.-2,6 linkages.
11) The method of claim 10, wherein the Fc-containing polypeptide has an N-
glycan
composition in which the amount and percentage of total sialylated N-glycans
is increased
relative to a parent Fc-containing polypeptide.
12) The method of claim 7, wherein the Fc-containing polypeptide is an
antibody or an
antibody fragment comprising sialylated N-glycans comprising a structure
selected from
SA(14)Gal(1-4)GleNAc(2-4)Man3GlcNAc2 or SAGalGlcNAcMan5GlcNAc2, wherein
the sialic acid residues are attached via a-2,6 linkages.
13) The method of claim 7, wherein the Fc-containing polypeptide has at least
one of the
following properties when compared to a parent Fc-containing polypeptide:
a) reduced effector function;
b) increased anti-inflammatory properties;
c) increased binding to a lectin (e.g., CD22 (Siglec 2));
d) reduced binding to Fc.gamma.RIIa;
e) increased binding to Fc.gamma.RIIb;
-71-

f) reduced binding to Fc.gamma.RIIIa; and
g) reduced binding to Fc.gamma.RIIIb.
14)A method of increasing the anti-inflammatory properties or decreasing
cytotoxicity of an
Fc-containing polypeptide comprising introducing mutations at positions 243,
264, 267
and 328 of the Fc region, wherein the numbering is according to the EU index
as in
Kabat;
wherein the Fc-containing polypeptide has increased anti-inflammatory
properties or
decreased cytotoxicity when compared to a parent Fc-containing polypeptide.
15) The method of claim 14, wherein the mutations are F243A, V264A, S267E, and
L328F.
16) The method of claim 14, werein the Fc-containing polypeptide comprises the
amino acid
sequence of SEQ ID NO:7, SEQ ID NO:8, or SEQ ID NO:17.
17) The method of claim 14, wherein the Fc-containing polypeptide is an
antibody or an
antibody fragment comprising sialylated N-glycans comprising a structure
selected from
SA(1-4)Gal(1-4)GlcNAc(2-4)Man3GlcNAc2 or SAGaIGICNAcMan5GlcNAc2 , wherein
the sialic acid residues are attached via .alpha.-2,6 linkages.
18) A method of treating an inflammatory condition in a subject in need
thereof comprising:
administering to the subject a therapeutically effective amount of an Fc-
containing
polypeptide comprising mutations at positions 243 and 264 of the Fc region,
wherein the
numbering is according to the EU index as in Kabat.
19) The method of claim 18, wherein the mutations are F243A, V264A, S267E, and
L328F.
20) The method of claim 18, werein the Fc-containing polypeptide comprises the
amino acid
sequence of SEQ ID NO:7, SEQ ID NO:8, or SEQ ID NO:17.
21) The method of claim 18, wherein the Fc-containing polypeptide is an
antibody or an
antibody fragment comprising sialylated N-glycans comprising a structure
selected from
-72-

SA(1-4)Gal(1-4)GlcNAC(2-4)Man3GlcNAc2 or SAGalGlcNAcMan5GlcNAc2 , wherein
the sialic acid residues are attached via .alpha.-2,6 linkages.
-73-

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
TITLE OF THE INVENTION
METHOD FOR PREPARING Fe-CONTAINING POLYPEPTIDES HAVING IMPROVED
PROPERTIES
FIELD OF THE INVENTION
The present invention is directed to methods and compositions for the
production of glycosylated proteins (glycoproteins) and, specifically, Fe-
containing
polypeptides which are useful as human or animal therapeutic agents.
BACKGROUND OF THE INVENTION
Monoclonal antibodies often achieve their therapeutic benefit through two
binding events. First, the variable domain of the antibody binds a specific
protein on a target
cell. This is followed by recruitment of effector cells that bind to the
constant region (Fe) of
the antibody and destroy cells to which the antibody is bound.
The potency of an antibody (or other immunotherapeutic composition)
depends on multiple mechanisms of action, including those mediated by effector
cells
expressing Fe receptors (FcRs). Fe receptors have activating or inhibitory
functional roles
and differ in their distribution among effector cells. Monocytes, macrophages,
and
neutrophils express both activating and inhibitory FcRs, whereas natural
killer (NK) cells
solely express the activating FcRIIIa. Thus, the degree to which an antibody
(or other
immunotherapeutic) can engage the various Fe receptors are important for
clinical outcome.
Amino acid- and glyco-engineering of the antibody Fe domain are two ways to
modify effector cell functions of antibodies and other immunothereapuetics.
See, e.g., Chu et
al., Molecular Immunology 45:3926-3933 (2008).
It would be desirable to engineer an antibody or Fe fusion protein comprising
improved properties. For example, it would be desirable to engineer antibodies
or other
immunotherapeutics which bind to Fe gamma receptor JIB (CD32B), but which do
not bind
(or binds with reduced affinity) to Fe gamma receptor IIA (CD32A) and Fe gamma
receptor
IIIA (CD16A) and Fe gamma receptor I (CD64). Such antibodies would be
characterized by
their lack of (or a significant decrease in) effector function and increased
anti-inflammatory
properties.
The presence of N-glycosylation not only plays a role in the effector function
of an antibody, the particular composition of the N-linked oligosaccharide is
also important
for its end function. For example, the lack of fucose or the presence of
bisecting N-acetyl
- 1 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
glucosamine has been positively correlated with the potency of the ADCC,
Rothman (1989),
Umana et al., Nat. Biotech. 17: 176-180 (1999), Shields et al., J. Biol. Chem.
277: 26733-
26740 (2002), and Shinkawa et al., J. Biol. Chem. 278: 3466-3473 (2003). There
is also
evidence that sialylation in the Fc region is positively correlated with the
anti-inflammatory
properties of intravenous immunoglobulin (IVIG). See, e.g., Kaneko et al.,
Science, 313:
670-673, 2006; Nimmerjahn and Ravetch, J. Exp. Med., 204: 11-15, 2007.
Given the utility of specific N-glycosylation in the function and potency of
antibodies, methods for modifying the composition of N-linked oligosaccharides
of
antibodies and method of modifying the effector functions of antibodies and
other
immunotherapeutics would be desirable.
Yeast and other fungal hosts are important production platforms for the
generation of recombinant proteins. Yeasts are eukaryotes and, therefore,
share common
evolutionary processes with higher eukaryotes, including many of the post-
translational
modifications that occur in the secretory pathway. Recent advances in
glycoengineering have
resulted in cell lines of the yeast strain Pichia pastoris with genetically
modified
glycosylation pathways that allow them to carry out a sequence of enzymatic
reactions, which
mimic the process of glycosylation in humans. See, for example, US Pat. Nos.
7,029,872,
7,326,681 and 7,449,308 that describe methods for producing a recombinant
glycoprotein in a
lower eukaryote host cell that are substantially identical to their human
counterparts. Human-
like sialylated bi-antennary complex N-linked glycans like those produced in
Pichia pastoris
from the aforesaid methods have demonstrated utility for the production of
therapeutic
glycoproteins. Thus, a method for further modifying or improving the
production of
antibodies in yeasts such as Pichia pastoris would be desirable.
SUMMARY OF THE INVENTION
The invention relates to an Fc-containing polypeptide comprising mutations at
amino acid positions 243, 264, 267 and 328 the Fc region, wherein the
numbering is
according to the EU index as in Kabat. In one embodiment, the mutations at
positions 243
are selected from the group consisting of: F243A, F243G, F243S, F243T, F243V,
F243L,
F243I, F243D, F243Y, F243E, F243R, F243W and F243K; the mutations at position
264 are
selected from the group consisting of: V264A, V264R, V264G, V264S, V264T,
V264D,
V264E, V264K, V264W, V264H, V264P, V264N, V264Q and V264L; the mutations at
position 267 are selected from the group consisiting of: S267D, S267Y, S267T;
and the
mutations at position 328 are selected from the group consisting of L328Y,
L328W, L328H.
- 2 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In one embodiment, the mutations at positions 243 and 264 are selected from
the group
consisting of: F243A and V264A; F243Y and V264G; F243T and V264G; F243L and
V264A; F243L and V264N; and F243V and V264G. In one embodiment, the mutations
are
F243A, V264A, S267E, and L328F.
In one embodiment, the Fe-containing polypeptide of the invention is an
antibody or an antibody fragment. In one embodiment, the Fe-containing
polypeptide of the
invention is an antibody fragment comprising SEQ ID NO:7, SEQ ID NO:8 or SEQ
ID
NO:17. In another embodiment, the Fe-containing polypeptide of the invention
is an
antibody fragment consisting (or consisting essentially of) SEQ ID NO:7, SEQ
ID NO:8 or
SEQ ID NO:17.
In one embodiment, the Fe-containing polypeptide is an antibody comprising
the heavy chain amino acid sequence of SEQ ID NO:4 or a variant thereof, and
the light chain
amino acid sequence of SEQ ID NO:2 or a variant thereof. In another
embodiment, theFc-
containing polypeptide is an antibody comprising the heavy chain amino acid
sequence of
SEQ ID NO:11 or a variant thereof, and the light chain amino acid sequence of
SEQ ID
NO:10 or a variant thereof In another embodiment, the Fe-containing
polypeptide is an
antibody comprising the heavy chain amino acid sequence of SEQ ID NO:12 or a
variant
thereof, and the light chain amino acid sequence of SEQ ID NO:13 or a variant
thereof.
In some embodiments, the Fe-containing polypeptides of the invention
comprise N-glycans comprising sialic acid (including NANA, NGNA, and analogs
and
derivatives thereof). In one embodiment, the N-glycans have a structure
selected from SA(1_
4)Gal(1-4)G1cNAc(2-4)Man3GleNAc2 or SAGa1G1eNAcMan5G1eNAc2. In one
embodiment, the Fe-containing polypeptides of the invention comprise a mixture
of a-2,3 and
a-2,6 linked sialic acid. In another embodiment, the Fc-contaning polypeptides
of the
invention comprise only a -2,6 linked sialic acid. In one embodiment, the Fc-
contaning
polypeptides of the invention comprise a -2,6 linked sialic acid and comprise
no detectable
level of a-2,3 linked sialic acid. In one embodiment, the sialic acid is N-
acetylneuraminic
acid (NANA) or N-glycolylneuraminic acid (NGNA) or a mixture thereof In
another
embodiment, the sialic acid is an analog or derivative of NANA or NGNA with
acetylation at
position 9 on the sialic acid. In one embodiment, the N-glycans on the Fe-
containing
polypeptides of the invention comprise NANA and no NGNA. In one embodiment,
the N-
glycans on the Fe-containing polypeptides of the invention comprise a -2,6
linked NANA
(and no NGNA). In one embodiment, the Fe-containing polypeptide of the
invention is an
- 3 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
antibody or an antibody fragment comprising sialylated N-glycans comprising a
structure
selected from SA(1_4)Gal(1_4)G1cNAc(2_4)Man3G1cNAc2 or SAGa1G1cNAcMan5G1cNAc2,
wherein the sialic acid residues are via a-2,6 linkages.
The N-glycans on the Fe-containing polypeptides of the invention can
optionally comprise
fucose. In one embodiment, the N-glycans on the Fe-containing polypeptides
will comprise a
mixture of fucosylated and non-fucosylated N-glycans. In another embodiment,
the N-
glycans on the Fe-containing polypeptides lack fucose.
In one embodiment, the Fe-containing polypeptide of the invention has one or
more of the following properties when compared to a parent Fe-containing
polypeptide: (i)
reduced effector function; (ii) increased anti-inflammatory properties; (iii)
increased binding
to a lectin (e.g., CD22 (Siglec 2)); (iv) reduced binding to FcyRIIa; (v)
increased binding to
FcyRIIb; (vi) reduced binding to FeyRIIIa; and (v) reduced binding to
FeyRIIIb.
In one embodiment, the Fe-containing polypeptide of the invention has
reduced effector function when compared to a parent Fe-containing polypeptide.
In one
embodiment, the effector function is ADCC. In another embodiment, the effector
function is
CDC. In another embodiment, the effector function is ADCP.
In one embodiment, the Fe-containing polypeptide of the invention has
reduced ADCC activity when compared to a parent Fe-containing polypeptide. In
another
embodiment, the Fe-containing polypeptide has at least a 100 fold reduction in
ADCC
activity. In another embodiment, the Fe-containing polypeptide has at least a
500 fold
reduction in ADCC activity. In another embodiment, the Fe-containing
polypeptide has at
least a 1000 fold reduction in ADCC activity. In one embodiment, the Fe-
containing
polypeptide has no detectable ADCC activity.
In another embodiment, the Fe-containing polypeptide of the invention has
reduced ADCP activity when compared to a parent Fe-containing polypeptide. In
one
embodiment, the Fe-containing polypeptide has no detectable ADCP activity.
In another embodiment, the Fe-containing polypeptide of the invention has
reduced CDC activity when compared to a parent Fe-containing polypeptide. In
one
embodiment, the Fe-containing polypeptide has at least 100 fold reduction in
CDC activity. In
one embodiment, the Fe-containing polypeptide has no detectable CDC activity.
In one embodiment, the Fe-containing polypeptide of the invention has the
following properties when compared to a parent Fe-containing polypeptide: (i)
reduced
binding to FeyRIIa; (ii) increased binding to FcyRIIb; (iii) reduced binding
to FeyRIIIa; and
(iv) reduced binding to FcyRIIIb.
- 4 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In one embodiment, the Fc-containing polypeptide of the invention has the
following properties when compared to a parent Fc-containing polypeptide: (i)
reduced
binding to Fc7RIIa; (ii) increased binding to FeyRlIb; and (iii) reduced
binding to FcyRIIIa.
In one embodiment, an Fe-containing polypeptide of the invention will have
no detectable binding to FeyRIIa, Fe7RIIIa or Fe7RIIIb. In one embodiment, an
Fe-containing
polypeptide of the invention will have no detectable binding to Fe7RIIa,
FcyRIIIa Fe7Rffib,
when such binding is detected using an ELISA assay.
In one embodiment, the Fc-containing polypeptide of the invention binds
FcyRlIb with an increase affinity of at least 2 fold when compared to a parent
Fe-containing
polypeptide. In one embodiment, the Fe-containing polypeptide of the invention
binds
FeyRlIb with an increase affinity of at least 4 fold when compared to a parent
Fe-containing
polypeptide.
In one embodiment, the Fe-containing polypeptide of the invention has
increased anti-inflammatory properties compared to a parent Fe-containing
polypeptide.
In a one embodiment, the parent Fe-containing polypeptide comprises a native
Fe region. In another embodiment, the parent Fe-containing polypeptide
comprises a F243A
mutation. In another embodiment, the parent Fe-containing polypeptide
comprises a V264A
mutation. In another embodiment, the parent Fe-containing polypeptide
comprises a F243A
mutation and a V264A mutation.
The invention also comprises a method for producing an Fe-containing
polypeptide in a host cell comprising: (i) providing a host cell that has been
genetically
engineered to produce an Fe-containing polypeptide, wherein the host cell
comprises a
nucleic acid encoding mutations at amino acid positions 243, 264, 267 and 328
of the Fe
region, wherein the numbering is according to the EU index as in Kabat; (ii)
culturing the
host cell under conditions which cause expression of the Fe-containing
polypeptide; and (iii)
isolating the Fe-containing polypeptide from the host cell. In one embodiment,
the nucleic
acid encodes mutations positions 243 are selected from the group consisting
of: F243A,
F243G, F243S, F243T, F243V, F243L, F243I, F243D, F243Y, F243E, F243R, F243W
and
F243K; the mutations at position 264 are selected from the group consisting
of: V264A,
V264R, V264G, V264S, V264T, V264D, V264E, V264K, V264W, V264H, V264P, V264N,
V264Q and V264L; the mutations at position 267 are selected from the group
consisiting of:
S267D, S267Y, S267T; and the mutations at position 328 are selected from the
group
consisting of L328Y, L328W, L328H. In one embodiment, the mutations at
positions 243
- 5 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
and 264 are selected from the group consisting of: F243A and V264A; F243Y and
V264G;
F243T and V264G; F243L and V264A; F243L and V264N; and F243V and V264G. In one
embodiment, the nucleic acid encodes the mutations F243A, V264A, S267E, and
L328F. In
one embodiment, the Fc-containing polypeptide of the invention is an antibody
or an antibody
fragment.
In one embodiment, the Fc-containing polypeptide of the invention is an
antibody fragment comprising SEQ ID NO:7, SEQ ID NO:8 or SEQ ID NO:17. In
another
embodiment, the Fc-containing polypeptide of the invention is an antibody
fragment
consisting (or consisting essentially of) SEQ ID NO:7, SEQ ID NO:8 or SEQ ID
NO:17.
In one embodiment, the method for producing an Fc-containing polypeptide is
carried out in a mammalian cell. In another embodiment, the method for
producing an Fc-
containing polypeptide is carried out in a plant cell. In another embodiment,
the method for
producing an Fc-containing polypeptide is carried out in bacteria. In another
embodiment, the
method for producing an Fc-containing polypeptide is carried out in an insect
cell. In another
embodiment, the method for producing an Fc-containing polypeptide is carried
out in a lower
eukaryotic cell. In another embodiment, the method for producing an Fc-
containing
polypeptide is carried out in a yeast cell. In one embodiment, the method for
producing an Fc-
containing polypeptide is carried out in Pichia pastoris.
In one embodiment, the Fc-containing polypeptide produced by the claimed
method comprises N-glycans comprising sialic acid (including NANA, NGNA, and
analogs
and derivatives thereof). In one embodiment, the Fc-containing polypeptide
produced by the
claimed method has an N-glycan composition in which at least 40 mole %, 70
mole % or 90
mole % of the N-glycans on the Fc-containing polypeptide are sialylated (have
a structure
selected from SA(1_4)Gal(1_4)G1cNAc(2_4)Man3G1cNAc2 or
SAGa1G1cNAcMan5G1cNAc2). In one embodiment, the Fc-containing polypeptides
produced by the claimed method comprise a mixture of a-2,3 and a-2,6 linked
sialic acid. In
another embodiment, the Fc-contaning polypeptides comprise only a-2,6 linked
sialic acid.
In one embodiment, the Fc-contaning polypeptides of the invention comprise a-
2,6 linked
sialic acid and comprise no detectable level of a-2,3 linked sialic acid. In
one embodiment,
the sialic acid is N-acetylneuraminic acid (NANA) or N-glycolylneuraminic acid
(NGNA) or
a mixture thereof. In another embodiment, the sialic acid is an analog or
derivative of NANA
or NGNA with acetylation at position 9 on the sialic acid. In one embodiment,
the N-glycans
on the Fc-containing polypeptides produced by the claimed method comprise NANA
and no
- 6 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
NGNA. In one embodiment, the N-glycans on the Fc-containing polypeptides of
the
invention comprise a -2,6 linked NANA (and no NGNA).
In one embodiment, the Fc-containing polypeptide of the invention is an
antibody or an antibody fragment comprising sialylated N-glycans comprising a
structure
selected from SA(1_4)Gal(1_4)G1cNAc(2_4)Man3G1cNAc2 or SAGa1G1eNAcMan5G1cNAc2,
wherein the sialic acid residues are via a-2,6 linkages.
The N-glycans on the Fc-containing polypeptides produced by the claimed
method can optionally comprise fucose. In one embodiment, the N-glycans on the
Fe-
containing polypeptides produced by the claimed method comprise a mixture of
fucosylated
and non-fucosylated N-glycans. In one embodiment, the N-glycans on the Fc-
containing
polypeptides produced by the claimed method lack fucose.
In one embodiment, the Fc-containing polypeptide produced by the claimed
method has an N-glycan composition in which the amount and percentage of total
sialylated
N-glycans is increased relative to a parent Fc-containing polypeptide.
In some embodiments, the Fc-containing polypeptide produced by the claimed
method has one or more of the following properties when compared to a parent
Fc-containing
polypeptide: (i) reduced effector function; (ii) increased anti-inflammatory
properties; (iii)
increased binding to a lectin (e.g., CD22 (Siglec 2)); (iv) reduced binding to
FcyRIIa; (v)
increased binding to FcyRIIb; (vi) reduced binding to FcyRIIIa; and (v)
reduced binding to
FcyRIIIb.
In one embodiment, the Fc-containing polypeptide produced by the claimed
method has reduced effector function when compared to a parent Fc-containing
polypeptide.
In one embodiment, the effector function is ADCC. In another embodiment, the
effector
function is CDC. In another embodiment, the effector function is ADCP.
In one embodiment, the Fe-containing polypeptide of the invention has
reduced ADCC activity when compared to a parent Fe-containing polypeptide. In
another
embodiment, the Fe-containing polypeptide has at least a 100 fold reduction in
ADCC
activity. In another embodiment, the Fe-containing polypeptide has at least a
500 fold
reduction in ADCC activity. In another embodiment, the Fe-containing
polypeptide has at
least a 1000 fold reduction in ADCC activity. In one embodiment, the Fe-
containing
polypeptide has no detectable ADCC activity.
In another embodiment, the Fe-containing polypeptide of the invention has
reduced ADCP activity when compared to a parent Fe-containing polypeptide. In
one
embodiment, the Fe-containing polypeptide has no detectable ADCP activity.
- 7 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In another embodiment, the Fe-containing polypeptide produced by the
claimed method has reduced CDC activity when compared to a parent Fe-
containing
polypeptide. In one embodiment, the Fe-containing polypeptide has at least 100
fold
reduction in CDC activity. In one embodiment, the Fe-containing polypeptide
has no
detectable CDC activity.
In one embodiment, the Fe-containing polypeptide produced by the claimed
method the following properties when compared to a parent Fe-containing
polypeptide: (i)
reduced binding to FcyRIIa; (ii) increased binding to FeyRnb; (iii) reduced
binding to
FcyRIIIa; and (v) reduced binding to FeyRIIIb.
In one embodiment, the Fe-containing polypeptide produced by the claimed
method the following properties when compared to a parent Fe-containing
polypeptide: (i)
reduced binding to FcyRIIa; (ii) increased binding to FcyRIIb; and (iii)
reduced binding to
FcyRIIIa.
In one embodiment, an Fe-containing polypeptide of the invention will have
no detectable binding to FeyRIIa, FcyRIIIa, or FeyRIIIb. In one embodiment, an
Fe-
containing polypeptide of the invention will have no detectable binding to
FcyRIIa, FcyRIIIa
or FeyRIIIb, when such binding is detected using an ELISA assay.
In one embodiment, the Fe-containing polypeptide of the invention binds
FeyRIIb with an increase affinity of at least 2 fold when compared to a parent
Fe-containing
polypeptide. In one embodiment, the Fe-containing polypeptide of the invention
binds
FcyRIIb with an increase affinity of at least 4 fold when compared to a parent
Fe-containing
polypeptide.
In one embodiment, the Fe-containing polypeptide produced by the claimed
method has increased anti-inflammatory properties relative to a parent Fe-
containing
polypeptide.
In a one embodiment, the parent Fe-containing polypeptide comprises a native
Fe region. In another embodiment, the parent Fe-containing polypeptide
comprises a F243A
mutation. In another embodiment, the parent Fe-containing polypeptide
comprises a V264A
mutation. In another embodiment, the parent Fe-containing polypeptide
comprises a F243A
mutation and a V264A mutation.
The invention also comprises a method of reducing the effector function of an
Fe-containing polypeptide, comprising introducing mutations at positions 243,
264, 267 and
328
- 8 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
of a parent Fc-contaning polypeptide, wherein said Fc containing polypeptide
has decreased
effector function when compared to the parent Fe-containing polypeptide,
wherein the
numbering is according to the EU index as in Kabat. In a one embodiment, the
Fe-containing
polypeptide comprises mutations F243A, V264A, S267E, and L328F. In one
embodiment,
the effector function is ADCC. In another embodiment, the effector function is
CDC. In one
embodiment, the effector function is ADCP. In one embodiment, the Fe-
containing
polypeptide of the invention is an antibody or an antibody fragment. In one
embodiment, the
Fe-containing polypeptide is an antibody fragment comprising SEQ ID NO:7. In
another
embodiment, the Fe-containing polypeptide is an antibody fragment comprising
SEQ ID
NO:8. In another embodiment, the Fe-containing polypeptide is an antibody
fragment
comprising SEQ ID NO:17. In another embodiment, the Fe-containing polypeptide
is an
antibody fragment consisting (or consisting essentially of) SEQ ID NO:7, SEQ
ID NO:8 or
SEQ ID NO:17. In one embodiment, the Fe-containing polypeptide of the
invention
comprises sialylated N-glycans comprising a structure selected from
SA(1_4)Gal( 1 _
4)GleNAc(2-4)Man3G1cNAc2 or SAGa1G1eNAcMan5G1cNAc2, wherein the sialic acid
residues are linked via a-2,6 linkages. In a one embodiment, the parent Fe-
containing
polypeptide comprises a native Fe region. In another embodiment, the parent Fe-
containing
polypeptide comprises a F243A mutation. In another embodiment, the parent Fe-
containing
polypeptide comprises a V264A mutation. In another embodiment, the parent Fe-
containing
polypeptide comprises a F243A mutation and a V264A mutation.
The invention also comprises a method of increasing the anti-inflammatory
properties of an Fe-containing polypeptide, comprising introducing mutations
at positions
243, 264, 267 and 328 of a parent Fe-contaning polypeptide, wherein the
numbering is
according to the EU index as in Kabat, wherein said Fe containing polypeptide
has increased
anti-inflammatory activity when compared to a parent Fe-containing
polypeptide. In a one
embodiment, the Fe-containing polypeptide comprises mutations F243A, V264A,
S267E, and
L328F. In one embodiment, the Fe-containing polypeptide of the invention is an
antibody or
an antibody fragment. In one embodiment, the Fe-containing polypeptide is an
antibody or
antigen binding fragment thereof that binds to an antigen selected from the
group consisting
of: APRIL, INF- a, BAFF (BLys), CD22, TNF-a, IL-1, IL-2, IL-4, IL-5, IL-6, IL-
8, IL-9, IL-
10, IL-12, IL-15, IL-17, IL-18, IL-20, IL-21, IL-22, IL-23, IL-23R, IL-25, IL-
27, IL-33, CD2,
CD4, CD11A, CD14, CD18, CD19, CD23, CD25, CD38, CD40, CD4OL, CD20, CD52,
- 9 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
CD64, CD80, CD147, CD200, CD200R, TSLP, TSLPR, PD-1, PDL1, CTLA4, VLA-4,
VEGF, PCSK9, a4f37-integrin, E-selectin, Fact II, ICAM-3, beta2-integrin,
IFI\17, C5, CBL,
LCAT, CR3, MDL-1, GITR, ADDL, CGRP, TRKA, IGF1R, RANKL, GTC, or the receptor
for any of the above mentioned molecules. In a one embodiment, the Fc-
containing
polypeptide will bind to TNF-a. In another embodiment, the Fc-containing
polypeptide will
bind to Her2. In another embodiment, the Fc-containing polypeptide will bind
to PCSK9. In
one embodiment, the Fc-containing polypeptide of the invention is an antibody
fragment
comprising SEQ ID NO:7. In another embodiment, the Fc-containing polypeptide
of the
invention is an antibody fragment comprising SEQ ID NO:8. In another
embodiment, the Fe-
containing polypeptide of the invention is an antibody fragment comprising SEQ
ID NO:17.
In another embodiment, the Fe-containing polypeptide of the invention is an
antibody
fragment consisting (or consisting essentially of) SEQ ID NO:7 or SEQ ID NO:8
or SEQ ID
NO:17. In one embodiment, the Fc-containing polypeptide of the invention is an
antibody or
an antibody fragment comprising sialylated N-glyeans comprising a structure
selected from
SA(1-4)Gal(1_4)G1cNAe(2_4)Man3G1cNAc2 or SAGa1G1eNAcMan5G1cNAc2, wherein the
sialic acid residues are linked via a-2,6 linkages. In one embodiment, the
parent Fc-
containing polypeptide comprises a native Fe region. In another embodiment,
the parent Fe-
containing polypeptide comprises a F243A mutation. In another embodiment, the
parent Fe-
containing polypeptide comprises a V264A mutation. In another embodiment, the
parent Fe-
containing polypeptide comprises a F243A mutation and a V264A mutation.
The invention also comprises a method of increasing the anti-inflammatory
properties of an Fe-containing polypeptide comprising: selecting a parent Fe-
containing
polypeptide that is useful in treating inflammation (for example, an antibody
or
immunoadhesin that binds to an antigen that is involved in inflammation) and
introducing
mutations at positions 243, 264, 267 and 328 of the Fe-region, wherein the
numbering is
according to the EU index as in Kabat, wherein the Fe-containing polypeptide
has increased
anti-inflammatory activity when compared to the parent Fe-containing
polypeptide. In one
embodiment, the nucleic acid encodes the mutations F243A, V264A, S267E, and
L328F. In
one embodiment, the Fe-containing polypeptide of the invention is an antibody
or an antibody
fragment. In one embodiment, the Fe-containing polypeptide is an antibody or
antigen
binding fragment thereof that binds to an antigen selected from the group
consisting of:
APRIL, INF- a, BAFF (BLys), CD22, TNF-a, IL-1, IL-2, IL-4, IL-5, IL-6, IL-8,
IL-9, IL-10,
IL-12, IL-15, IL-17, IL-18, IL-20, IL-21, IL-22, IL-23, IL-23R, IL-25, IL-27,
IL-33, CD2,
-10-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
CD4, CD11A, CD14, CD18, CD19, CD23, CD25, CD38, CD40, CD4OL, CD20, CD52,
CD64, CD80, CD147, CD200, CD200R, TSLP, TSLPR, PD-1, PDL1, CTLA4, VLA-4,
VEGF, PCSK9, a407-integrin, E-selectin, Fact II, ICAM-3, beta2-integrin, IFNy,
C5, CBL,
LCAT, CR3, MDL-1, GITR, ADDL, CGRP, TRKA, IGF1R, RANKL, GTC, or the receptor
for any of the above mentioned molecules. In a one embodiment, the Fe-
containing
polypeptide will bind to TNF-a. In another one embodiment, the Fe-containing
polypeptide
will bind to Her2. In another one embodiment, the Fe-containing polypeptide
will bind to
PCSK9. In one embodiment, the Fe-containing polypeptide is an antibody
fragment
comprising SEQ ID NO:7. In another embodiment, the Fe-containing polypeptide
is an
antibody fragment comprising SEQ ID NO:8. In another embodiment, the Fe-
containing
polypeptide of the invention is an antibody fragment comprising SEQ ID NO:17.
In another
embodiment, the Fe-containing polypeptide is an antibody fragment consisting
(or consisting
essentially of) SEQ ID NO:7 or SEQ ID NO:8 or SEQ ID NO:17. In one embodiment,
the
Fe-containing polypeptide of the invention is an antibody or an antibody
fragment comprising
sialylated N-glycans comprising a structure selected from
SA(1_4)Gal(1_4)G1cNAc(2-
4)Man3G1cNAc2 or SAGa1G1eNAcMan5G1cNAc2, wherein the sialic acid residues are
linked via a-2,6 linkages. In one embodiment, the parent Fe-containing
polypeptide
comprises a native Fe region. In another embodiment, the parent Fe-containing
polypeptide
comprises a F243A mutation. In another embodiment, the parent Fe-containing
polypeptide
comprises a V264A mutation. In another embodiment, the parent Fe-containing
polypeptide
comprises a F243A mutation and a V264A mutation.
The invention also comprises a method of treating an inflammatory condition
in a subject in need thereof comprising: administering to the subject a
therapeutically
effective amount of an Fe-containing polypeptide comprising mutations at
positions 243, 264,
267 and 328, wherein the numbering is according to the EU index as in Kabat.
In one
embodiment, the Fe-containing polypeptide decreases the expression of a gene
selected from
the group consisting of: IL-10, IL-6, RANKL, TRAP, ATP6v0d2, MDL-1, DAP12,
CD11b,
TIMP-1, MMP9, CTSK, PU-1, MCP1, MIPla, Cxcll-Groa, CXc12-Grob, CD18, TNF,
FcyRI, FcyRlIb, FcyRIII and FcyRIV. In a one embodiment, the Fe-containing
polypeptide
comprises mutations F243A, V264A, S267E, and L328F. In one embodiment, the Fe-
containing polypeptide is administered parenterally. In one embodiment, the Fe-
containing
polypeptide is administered subcutaneously. In one embodiment, the Fe-
containing
-11-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
polypeptide is an antibody or antigen binding fragment thereof. In one
embodiment, the Fe-
containing polypeptide is an antibody or antigen binding fragment thereof that
is useful in
treating an inflammatory condition. In one embodiment, the antibody or antigen
binding
fragment thereof binds to an antigen selected from the group consisting of:
APRIL, INF- a,
BAFF (BLys), CD22, TNF-a, IL-1, IL-2, IL-4, IL-5, IL-6, IL-8, IL-9, IL-10, IL-
12, IL-15, IL-
17, IL-18, IL-20, IL-21, IL-22, IL-23, IL-23R, IL-25, IL-27, IL-33, CD2, CD4,
CD11A,
CD14, CD18, CD19, CD23, CD25, CD38, CD40, CD4OL, CD20, CD52, CD64, CD80,
CD147, CD200, CD200R, TSLP, TSLPR, PD-1, PDL1, CTLA4, VLA-4, VEGF, PCSK9,
a4137-integrin, E-selectin, Fact II, ICAM-3, beta2-integrin, IFN7, C5, CBL,
LCAT, CR3,
MDL-1, GITR, ADDL, CGRP, TRKA, IGFIR, RANKL, GTC, or the receptor for any of
the
above mentioned molecules. In one embodiment, the Fc-containing polypeptide
will bind to
TNF-a. In another embodiment, the Fc-containing polypeptide will bind to Her2.
In another
embodiment, the Fc-containing polypeptide will bind to PCSK9. In one
embodiment, the Fe-
containing polypeptide is an antibody fragment comprising SEQ ID NO:7. In
another
embodiment, the Fc-containing polypeptide is an antibody fragment comprising
SEQ ID
NO:8. In another embodiment, the Fc-containing polypeptide of the invention is
an antibody
fragment comprising SEQ ID NO:17. In another embodiment, the Fc-containing
polypeptide
is an antibody fragment consisting (or consisting essentially of) SEQ ID NO:7
or SEQ ID
NO:8 or SEQ ID NO:17. In one embodiment, the Fc-containing polypeptide of the
invention
is an antibody or an antibody fragment comprising sialylated N-glycans
comprising a
structure selected from SA(1-4)Gal(1-4)G1cNAc(2-4)Man3G1cNAc2 or
SAGa1G1cNAcMan5G1cNAc2, wherein the sialic acid residues are linked via a-2,6
linkages.
Another invention disclosed herein relates to a pharmaceutical composition
comprising an Fc-containing polypeptide, wherein at least 70%, at least 80% or
at least 90%
of the N-glycans on the Fc-containing polypeptide comprise an oligosaccharide
structure
selected from the group consisting of SA(1_4.)Gal(i..4)G1cNAc(2_4)Man3G1cNAc2
and
SAGa1G1eNAcMan5G1cNAc2, wherein the Fc-containing polypeptide comprises
mutations at
amino acid positions 243, 264, 267 and 328 of the Fc region, wherein the
numbering is
according to the EU index as in Kabat. In one embodiment, the mutations are
F243A,
V264A, S267E, and L328F. In one embodiment, the sialylated N-glycans comprise
a mixture
of a-2,3 and a-2,6 linked sialic acid. In another embodiment, the sialylated N-
glycans
comprise only a-2,6 linked sialic acid. In another embodiment, the sialylated
N-glycans
- 12 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
comprise a-2,6 linked sialic acid and comprise no detectable level of a-2,3
linked sialic acid.
In one embodiment, the sialic acid is N-acetylneuraminic acid (NANA) or N-
glycolylneuraminic acid (NGNA) or a mixture thereof. In another embodiment,
the sialic
acid is an analog or derivative of NANA or NGNA with acetylation at position 9
on the sialic
acid. In one embodiment, the N-glycans on the Fe-containing polypeptides
comprise NANA
and no NGNA. In one embodiment, the Fe-containing polypeptide is an antibody
fragment
comprising SEQ ID NO:7. In another embodiment, the Fe-containing polypeptide
is an
antibody fragment comprising SEQ ID NO:8. In another embodiment, the Fe-
containing
polypeptide of the invention is an antibody fragment comprising SEQ ID NO:17.
In another
embodiment, the Fe-containing polypeptide is an antibody fragment consisting
(or consisting
essentially of) SEQ ID NO:7 or SEQ ID NO:8 or SEQ ID NO:17. In one embodiment,
the
Fe-containing polypeptide of the invention is an antibody or an antibody
fragment comprising
sialylated N-glycans comprising a structure selected from SA(1_4)Gal(1
_4)G1cNAc(2_
4)Man3G1cNAc2 or SAGa1G1eNAcMan5G1cNAc2, wherein the sialic acid residues are
linked via a-2,6 linkages.
Another invention disclosed herein relates to a pharmaceutical composition
comprising an Fe-containing polypeptide, wherein at least 70%, 80% or 90% of
the N-glycans
on the Fe-containing polypeptide comprise an oligosaccharide structure
selected from the
group consisting of SA(1_4)Gal(1 _4)GleNAc(2_4)Man3G1cNAc2 and
SAGa1G1eNAcMan5G1cNAc2, wherein the sialic acid residues are attached
exclusively via
an a -2,6 linkage, wherein the N-glycans lack fucose, and wherein the Fe-
containing
polypeptide comprises mutations at amino acid positions 243, 264, 267 and 328
of the Fe
region, wherein the numbering is according to the EU index as in Kabat. In one
embodiment,
the mutations are F243A, V264A, S267E, and L328F. In one embodiment, the
sialylated N-
glycans comprise a mixture of a-2,3 and a-2,6 linked sialic acid. In another
embodiment, the
sialylated N-glycans comprise only a -2,6 linked sialic acid. In another
embodiment, the
sialylated N-glycans comprise a-2,6 linked sialic acid and comprise no
detectable level of a-
2,3 linked sialic acid. In one embodiment, the sialic acid is N-
acetylneuraminic acid (NANA)
or N-glycolylneuraminic acid (NGNA) or a mixture thereof In another
embodiment, the
sialic acid is an analog or derivative of NANA or NGNA with acetylation at
position 9 on the
sialic acid. In one embodiment, the N-glycans on the Fe-containing
polypeptides comprise
NANA and no NGNA. In one embodiment, the N-glycans on the Fe-containing
polypeptides
- 13 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
of the invention comprise a -2,6 linked NANA (and no NGNA). In one embodiment,
the Fe-
containing polypeptide is an antibody fragment comprising SEQ ID NO:7. In
another
embodiment, the Fc-containing polypeptide is an antibody fragment comprising
SEQ ID
NO:8. In another embodiment, the Fe-containing polypeptide of the invention is
an antibody
fragment comprising SEQ ID NO:17. In another embodiment, the Fe-containing
polypeptide
is an antibody fragment consisting (or consisting essentially of) SEQ ID NO:7
or SEQ ID
NO:8 or SEQ ID NO:17.
The invention also comprises an Fe-containing polypeptide comprising a
heavy chain and a light chain, wherein the heavy chain comprises the amino
acid sequence of
SEQ ID NO:4 or a variant thereof and the light chain comprises the amino acid
sequence of
SEQ ID NO:2 or a variant thereof, wherein the variant comprises one or more of
the
following properties when compared to an antibody comprising the heavy chain
amino acid
sequence of SEQ ID NO:1 and the light chain amino acid sequence of SEQ ID
NO:2: (i)
reduced effector function; (ii) increased anti-inflammatory properties; (iii)
increased binding
to a lectin (e.g., CD22 (Siglec 2)); (iv) reduced binding to FeyRIIa; (v)
increased binding to
FcyRIIb; (vi) reduced binding to FeyRIIIa; and (vii) reduced binding to
FcyRIIIb. In one
embodiment, the variant comprises up to 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or
more conservative or
non conservative amino acid substitutions. In one embodiment, the variant
comprises at least
75%, 80%, 85%, 90%, 95%, 98% or 99% sequence identity with the claimed
sequence.
The invention also comprises an Fe-containing polypeptide comprising a
heavy chain and a light chain, wherein the heavy chain comprises the amino
acid sequence of
SEQ ID NO:4 or a variant thereof and the light chain comprises the amino acid
sequence of
SEQ ID NO:2 or a variant thereof, wherein the variant comprises one or more of
the
following properties when compared to an antibody comprising the heavy chain
amino acid
sequence of SEQ ID NO:3 and the light chain amino acid sequence of SEQ ID
NO:2: (i)
reduced effector function; (ii) increased anti-inflammatory properties; (iii)
increased binding
to a lectin (e.g., CD22 (Siglec 2)); (iv) reduced binding to FcyRIIa; (v)
increased binding to
FcyRIIb; (vi) reduced binding to FcyRIIIa; and (vii) reduced binding to
FcyRIIIb. In one
embodiment, the variant comprises up to 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or
more conservative or
non conservative amino acid substitutions. In one embodiment, the variant
comprises at least
75%, 80%, 85%, 90%, 95%, 98% or 99% sequence identity with the claimed
sequence.
- 14 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graphic representation of pGLY8068, the expression plasmid for
antibody 1F11 F243A/V264A/S267E/L328F. Both heavy and light chains were under
the
control of a methanol inducible promoter, A0X1. The PpTrp2 gene was the locus
applied to
Figure 2 is a representation of the gels from an SDS-PAGE analysis
Figures 3-8 are graphic representations of the FcyR binding properties of the
Fc-containing polypeptides of the invention.
Figure 9 shows the effect of some of the Fc polypeptides of the invention in a
murine ITP model.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
The term "GO" when used herein refers to a complex bi-antennary
oligosaccharide without galactose or fucose, GlcNAc2Man3G1cNAc2.
The term "Gl" when used herein refers to a complex bi-antennary
oligosaccharide without fucose and containing one galactosyl residue,
GalG1cNAc2Man3G1cNAc2.
The term "G2" when used herein refers to a complex bi-antennary
oligosaccharide without fucose and containing two galactosyl residues,
Gal2G1cNAc2Man3G1cNAc2.
The term "GOF" when used herein refers to a complex bi-antennary
oligosaccharide containing a core fucose and without galactose,
G1cNAc2Man3G1cNAc2F.
- 15 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
The term "GlF" when used herein refers to a complex bi-antennary
oligosaccharide containing a core fucose and one galactosyl residue,
GalGleNAc2Man3G1cNAc2F.
The term "G2F" when used herein refers to a complex bi-antennary
oligosaccharide containing a core fucose and two galactosyl residues,
Gal2G1cNAc2Man3G1cNAc2F.
The term "Man5" when used herein refers to the oligosaccharide structure
shown as
_40.44 a1,3 Mannose
= G1cNAc
1= 31,4a1,3
= a1,6 Mannose
The term "M4" as used herein refers to the oligosaccharide Man4G1cNAc2.
The term "M5" as used herein refers to the oligosaccharide Man5G1cNAc2.
The term "Al" as used herein refers to the oligosaccharide
NANAlGal2G1cNAc2Man3G1cNAc2.
The term "Al H" as used herein refers to the oligosaccharide
NANA1GalMan(3 -5)G1cNAc2.
The term "A2" as used herein refers to the oligosaccharide
NANA2Ga12G1cNAc2Man3G1cNAc2.
The term "GFI 5.0" when used herein refers to glycoengineered Pichia
pastoris strains that produce glycoproteins having predominantly
Gal2G1cNAc2Man3G1cNAc2 N-glycans.
The term "GFI 6.0" when used herein refers to glycoengineered Pichia
pastoris strains that produce glycoproteins having predominantly
NANA2Ga12G1cNAc2Man3G1cNAc2 N-glycans.
The term "GS5.0", when used herein refers to the N-glycosylation structure
Gal2G1cNAc2Man3G1cNAc2.
The term "GS5.5", when used herein refers to the N-glycosylation structure
NANAGa12G1cNAc2Man3G1cNAc2, which when produced in Pichia pastoris strains to
which a 2,6 sialyl transferase has been glycoengineered result in a 2,6-linked
sialic acid and
- 16 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
which when produced in Pichia pastoris strains to which a 2,3 sialyl
transferase has been
glycoengineered result in a 2,3-linked sialic acid.
The term "GS6.0", when used herein refers to the N-glycosylation structure
NANA2Ga12G1cNAc2Man3G1cNAc2, which when produced in Pichia pastoris strains to
which a 2,6 sialyl transferase has been glycoengineered result in a 2,6-linked
sialic acid and
which when produced in Pichia pastoris strains to which a 2,3 sialyl
transferase has been
glycoengineered result in a 2,3-linked sialic acid.
The term "wild type" or "wt" when used herein in connection to a Pichia
pastoris strain refers to a native Pichia pastoris strain that has not been
subjected to genetic
modification to control glycosylation.
The term "antibody", when used herein refers to an immunoglobulin molecule
capable of binding to a specific antigen through at least one antigen
recognition site located in
the variable region of the immunoglobulin molecule. As used herein, the term
encompasses
not only intact polyclonal or monoclonal antibodies, consisting of four
polypeptide chains, i.e.
two identical pairs of polypeptide chains, each pair having one "light" chain
(LC) (about 25
kDa) and one "heavy" chain (HC) (about 50-70 kDa), but also fragments
thereof', such as Fab,
Fab', F(a131)2, Fv, single chain (ScFv), mutants thereof, fusion proteins
comprising an
antibody portion, and any other modified configuration of an immunoglobulin
molecule that
comprises an antigen recognition site and at least the portion of the CH2
domain of the heavy
chain immunoglobulin constant region which comprises an N-linked glycosylation
site of the
CH2 domain, or a variant thereof As used herein the term includes an antibody
of any class,
such as IgG (for example, IgGl, IgG2, IgG3 or IgG4), IgM, IgA, IgD and IgE,
respectively.
The term "consensus sequence of CH2" when used herein refers to the amino
acid sequence of the CH2 domain of the heavy chain constant region containing
an N-linked
glycosylation site which was derived from the most common amino acid sequences
found in
CH2 domains from a variety of antibodies.
The term "Fe region" is used to define a C-terminal region of an
immunoglobulin heavy chain. The "Fe region" may be a native sequence Fe region
or a
variant Fe region. Although the boundaries of the Fe region of an
immunoglobulin heavy
chain might vary, the human IgG heavy chain Fe region is usually defined to
stretch from an
amino acid residue at position Cys226, or from Pro230, to the carboxyl-
terminus thereof The
Fe region of an immunoglobulin comprises two constant domains, CH2 and CH3,
and can
optionally comprise a hinge region. In one embodiment, the Fe region comprises
the amino
- 17 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
acid sequence of SEQ ID NO:7. In one embodiment, the Fc region comprises the
amino acid
sequence of SEQ ID NO:8. In another embodiment, the Fc-containing polypeptide
of the
invention is an antibody fragment comprising SEQ ID NO:17. In another
embodiment, the Fc
region comprises the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8, with
the
addition of a lysine (K) residue at the 3' end. The Fc region contains a
single N-linked
glycosylation site in the CH2 domain that corresponds to the Asn297 site of a
full-length
heavy chain of an antibody.
The term "Fc-containing polypeptide" refers to a polypeptide, such as an
antibody or immunoadhesin, which comprises an Fc region. This term encompasses
polypeptides comprising or consisting of (or consisting essentially of) an Fc
region.
Polypeptides comprising an Fc region can be generated by papain digestion of
antibodies or
by recombinant DNA technology.
The term "parent antibody", "parent immunoglobulin" or "parent Fc-
containing polypeptide" when used herein refers to an antibody or Fc-
containing polypeptide
which lacks the Fc region mutations disclosed herein. A parent Fc-containing
polypeptide
may comprise a native sequence Fc region or an Fc region with pre-existing
amino acid
sequence modifications. A native sequence Fc region comprises an amino acid
sequence
identical to the amino acid sequence of an Fc region found in nature. Native
sequence Fc
regions include the native sequence human IgG1 Fc region, the native sequence
human IgG2
Fc region, the native sequence human IgG3 Fc region and the native sequence
human IgG4 Fc
region as well as naturally occurring variants thereof. When used as a
comparator, a parent
antibody or a parent Fc-containing polypeptide can be expressed in any cell.
In one
embodiment, the parent antibody or a parent Fc-containing polypeptide is
expressed in the
same cell as the Fc-containing polypeptide of the invention.
As used herein, the term "immunoadhesin" designates antibody-like molecules
which combine the "binding domain" of a heterologous "adhesin" protein (e.g. a
receptor,
ligand or enzyme) with an immunoglobulin constant domain. Structurally, the
immunoadhesins comprise a fusion of the adhesin amino acid sequence with the
desired
binding specificity which is other than the antigen recognition and binding
site (antigen
combining site) of an antibody (i.e. is "heterologous") and an immunoglobulin
constant
domain sequence. The term "ligand binding domain" as used herein refers to any
native cell-
surface receptor or any region or derivative thereof retaining at least a
qualitative ligand
binding ability of a corresponding native receptor. In a specific embodiment,
the receptor is
from a cell-surface polypeptide having an extracellular domain that is
homologous to a
- 18 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
member of the immunoglobulin supergenefamily. Other receptors, which are not
members of
the immunoglobulin supergenefamily but are nonetheless specifically covered by
this
definition, are receptors for cytokines, and in particular receptors with
tyrosine kinase activity
(receptor tyrosine kinases), members of the hematopoietin and nerve growth
factor which
predispose the mammal to the disorder in question. In one embodiment, the
disorder is
cancer. Methods of making immunoadhesins are well known in the art. See, e.g.,
W000/42072.
The antibody referred to as "1F11" refers to a humanized anti-PCSK9
antibody having the amino acid sequence disclosed in Example 2.
The term "Herceptin0" refers to the commercial anti-Her2 antibody produced
in CHO cells also known as rastuzumab.
The term "Fc mutein antibody" when used herein refers to an antibody
comprising one of the single Fc muteins or the double Fc mutein described
herein.
The term "Fc mutein" when used herein refers to an Fc-containing polypeptide
in which one or more point mutations have been made to the Fc region.
The term "Fc mutation" when used herein refers to a mutation made to the Fc
region of an Fc-containing polypeptide. Examples of such a mutation include
the F243A,
V264A, S267E, or L328F mutations described herein.
The term "F243A" refers to a mutation from F (wild-type) to A at position 243
The term "double Fe mutein" or "DM" when used herein refers to an Fc-
containing polypeptide comprising mutations at positions 243 and 264 of the Fe
region. The
term "F243A/V264A" refers to a double Fc mutein comprising the two specified
mutations.
30 The term "quadruple Fc mutein" or "QM" when used herein refers to
an Fc-
containing polypeptide comprising mutations at positions 243, 264, 267 and 328
of the Fe
region of the antibody heavy chain. The term "F243A/V264A/S267E/L328F" refers
to a
quadruple Fc mutein comprising the four specified mutations.
- 19 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Throughout the present specification and claims, the numbering of the residues
in an immunoglobulin heavy chain or an Fc-containing polypeptide is that of
the EU index as
in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
Public Health
Service, National Institutes of Health, Bethesda, MD (1991), expressly
incorporated herein by
reference. The "EU index as in Kabat" refers to the residue numbering of the
human IgG1 EU
antibody.
The term "effector function" as used herein refers to a biochemical event that
results from the interaction of an antibody Fc region with an Fc receptor or
ligand.
Exemplary "effector functions" include Clq binding; complement dependent
cytotoxicity; Fc
receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC);
phagocytosis;
down regulation of cell surface receptors (e. g. B cell receptor; BCR), etc.
Such effector
functions can be assessed using various assays known in the art.
The term "glycoengineered Pichia pastoris" when used herein refers to a strain
of Pichia pastoris that has been genetically altered to express human-like N-
glycans. For
example, the GFI 5.0, GFI 5.5 and GFI 6.0 strains described above.
The terms "N-glycan", "glycoprotein" and "glycoform" when used herein refer
to an N-linked oligosaccharide, e.g., one that is attached by an asparagine-N-
acetylglucosamine linkage to an asparagine residue of a polypeptide.
Predominant sugars
found on glycoproteins are glucose, galactose, mannose, fucose, N-
acetylgalactosamine
(GalNAc), N-acetylglucosamine (GleNAc) and sialic acid (SA, including NANA,
NGNA and
derivatives and analogs thereof, including acetylated NANA or acetylated
NGNA). In
glycoengineered Pichia pastoris, sialic acid is exclusively N-acetyl-
neuraminic acid (NANA)
(Hamilton et al., Science 313 (5792): 1441-1443 (2006)). N-glycans have a
common
pentasaccharide core of Man3G1cNAc2, wherein "Man" refers to mannose, "Glc"
refers to
glucose, "NAc" refers to N-acetyl, and GleNAc refers to N-acetylglucosamine. N-
glycans
differ with respect to the number of branches (antennae) comprising peripheral
sugars (e.g.,
GlcNAc, galactose, fucose and sialic acid) that are added to the Man3G1cNAc2
("Man3")
core structure which is also referred to as the "trimannose core", the
"pentasaccharide core"
or the "paucimannose core". N-glycans are classified according to their
branched constituents
(e.g., high mannose, complex or hybrid).
As used herein, the term "sialic acid" or "SA" refers to any member of the
sialic acid family, including without limitation: N-acetylneuraminic acid
(Neu5Ac or
NANA), N-glycolylneuraminic acid (NGNA) and any analog or derivative thereof
(including
- 20 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
those arising from acetylation at any position on the sialic acid molecule).
Sialic acid is a
generic name for a group of about 30 naturally occurring acidic carbohydrates
that are
essential components of a large number of glycoconjugates. Schauer, Biochem.
Society
Transactions, 11, 270-271 (1983). Sialic acids are usually the terminal
residue of the
oligosaccharides. N-acetylneuraminic acid (NANA) is the most common sialic
acid form and
N-glycolylneuraminic acid (NGNA) is the second most common form. Schauer,
Glycobiolog3T, 1, 449-452 (1991). NGNA is widespread throughout the animal
kingdom and,
according to species and tissue, often constitutes a significant proportion of
the
glycoconjugate-bound sialic acid. Certain species such as chicken and man are
exceptional,
since they lack NGNA in normal tissues. Corfield, et al., Cell Biology
Monographs, 10, 5-50
(1982). In human serum samples, the percentage of sialic acid in the form of
NGNA is
reported to be 0.01% of the total sialic acid. Schauer, "Sialic Acids as
Antigenic Determinants
of Complex Carbohydrates", found in The Molecular Immunology of Complex
Carbohydrates, (Plenum Press, New York, 1988).
The term "human-like N-glycan", as used herein, refers to the N-linked
oligosaccharides which closely resemble the oligosaccharides produced by non-
engineered,
wild-type human cells. For example, wild-type Pichia pastoris and other lower
eukaryotic
cells typically produce hypermannosylated proteins at N-glycosylation sites.
The host cells
described herein produce glycoproteins (for example, antibodies) comprising
human-like N-
glycans that are not hypermannosylated. In some embodiments, the host cells of
the present
invention are capable of producing human-like N-glycans with hybrid and/or
complex N-
glycans. The specific type of "human-like" glycans present on a specific
glycoprotein
produced from a host cell of the invention will depend upon the specific
glycoengineering
steps that are performed in the host cell.
The term "high mannose" type N-glycan when used herein refers to an N-
glycan having five or more mannose residues.
The term "complex" type N-glycan when used herein refers to an N-glycan
having at least one GlcNAc attached to the 1,3 mannose arm and at least one
GlcNAc
attached to the 1,6 mannose arm of a "trimannose" core. Complex N-glycans may
also have
galactose ("Gal") or N-acetylgalactosamine ("GalNAc") residues that are
optionally modified
with sialic acid or derivatives (e.g., "NANA" or "NeuAc", where "Neu" refers
to neuraminic
acid and "Ac" refers to acetyl). Complex N-glycans may also have intrachain
substitutions
comprising "bisecting" GlcNAc and core fucose ("Fuc"). As an example, when a N-
glycan
comprises a bisecting GleNAc on the trimannose core, the structure can be
represented as
-21-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Man3G1cNAc2(G1cNAc) or Man3G1cNAc3. When an N-glycan comprises a core fucose
attached to the trimannose core, the structure may be represented as
Man3G1cNAc2(Fuc).
Complex N-glycans may also have multiple antennae on the "trimannose core,"
often referred
to as "multiple antennary glycans."
The term "hybrid" N-glycan when used herein refers to an N-glycan having at
least one GleNAc on the terminal of the 1,3 mannose arm of the trimannose core
and zero or
more than one mannose on the 1,6 mannose arm of the trimannose core.
When referring to "mole percent" of a glycan present in a preparation of a
glycoprotein, the term means the molar percent of a particular glycan present
in the pool of N-
linked oligosaccharides released when the protein preparation is treated with
PNGase and
then quantified by a method that is not affected by glycoform composition,
(for instance,
labeling a PNGase released glycan pool with a fluorescent tag such as 2-
aminobenzamide and
then separating by high performance liquid chromatography or capillary
electrophoresis and
then quantifying glycans by fluorescence intensity). For example, 50 mole
percent NANA2
Ga12G1cNAc2Man3G1cNAc2 means that 50 percent of the released glycans are NANA2
Gal2G1cNAc2Man3G1cNAc2 and the remaining 50 percent are comprised of other N-
linked
oligosaccharides.
The term "anti-inflammatory antibody" as used herein, refers to an antibody
intended to be used to treat inflammation. The anti-inflammatory properties of
an Fc-
containing polypeptide can be measured using any method known in the art. In
one
embodiment, the anti-inflammatory properties of an Fc-containing polypeptide
are measured
using an animal model, such as the models described in Kaneko et al., Science
313:670-673
(2006), Anthony et al., Science 320:373-376 (2008), and Examples 20-21 herein.
In another
embodiment, the anti-inflammatory properties of an Fc-containing polypeptide
are measured
by determining the level of a biomarker related to inflammation (including
without limitation:
CRP, pro-inflammatory cytokines such as tumor necrosis factors (TNF-alpha),
interferon-
gamma, interleukin 6 (IL-6, IL-8, IL-10, chemokines, the coagulation marker D-
dimer,
sCD14, intestinal fatty acid binding peptide (IFABP), and hyaluronic acid. In
one
embodiment, the anti-inflammatory properties of an Fc-containing polypetpide
is measured
by determining the level of C-reactive protein (CRP) using a method known in
the art. A
decrease in the level of C-reactive protein indicates that the Fc-containing
polypeptide has
anti-inflammatory properties.
- 22 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
"Conservatively modified variants" or "conservative substitution" refers to
substitutions of amino acids in a protein with other amino acids having
similar characteristics
(e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone
conformation and
rigidity, etc.), such that the changes can frequently be made without altering
the biological
activity of the protein. Those of skill in this art recognize that, in
general, single amino acid
substitutions in non-essential regions of a polypeptide do not substantially
alter biological
activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The
Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of
structurally or
functionally similar amino acids are less likely to disrupt biological
activity. Exemplary
conservative substitutions are listed below:
Original residue Conservative substitution
Ala (A) Gly; Ser
Arg (R) Lys; His
Asn (N) Gln; His
Asp (D) Glu; Asn
Cys (C) Ser; Ala
Gln (Q) Asn
Glu (E) Asp; Gln
Gly (G) Ala
His (H) Asn; Gln
Ile (I) Leu; Val
Leu (L) Ile; Val
Lys (K) Arg; His
Met (M) Leu; Ile; Tyr
Phe (F) Tyr; Met; Leu
Pro (P) Ala
Ser (S) Thr
Thr (T) Ser
Trp (W) Tyr; Phe
Tyr (Y) Trp; Phe
Val (V) Ile; Leu
Glycosylation of immunoglobulin G (IgG) in the Fc region, Asn297 (according
to the EU numbering system), has been shown to be a requirement for optimal
recognition
and activation of effector pathways including antibody dependent cellular
cytotoxicity
- 23 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
in immune effector function assays including ADCC, Rothman (1989), Lifely et
al.,
Glycobiology, 5:813-822 (1995), Umana (1999), Shields (2002), and Shinkawa
(2003), and
complement dependent cytotoxicity (CDC), Hodoniczky et al., Biotechnol. Prog.,
21(6):
1644-1652 (2005), and Jefferis et al., Chem. Immunol., 65: 111-128 (1997).
This effect on
function has been attributed to the specific conformation adopted by the
glycosylated Fc
domain, which appears to be lacking when glycosylation is absent. More
specifically, IgG
which lacks glycosylation in the Fc CH2 domain does not bind to Fc7R,
including Fc7RI,
Fc7RII, and Fc7RIII, Rothman (1989).
Not only does the presence of glycosylation appear to play a role in the
effector function of an antibody, the particular composition of the N-linked
oligosaccharide is
also important. For example, the presence of fucose shows a marked effect on
in vitro
Fc7RIIIa binding and in vitro ADCC, Rothman (1989), and Li et al., Nat.
Biotechnol. 24(2):
2100-215 (2006). Recombinant antibodies produced by mammalian cell culture,
such as
CHO or NSO, contain N-linked oligosaccharides that are predominately
fucosylated, Hossler
etal., Biotechnology and Bioengineering, 95(5):946-960 (2006), Umana (1999),
and Jefferis
et al., Biotechnol. Prog. 21:11-16 (2005). Additionally, there is evidence
that sialylation in
the Fc region may impart anti-inflammatory properties to antibodies.
Intravenous
immunoglobulin (IVIG) purified over a lectin column to enrich for the
sialylated form
showed a distinct anti-inflammatory effect limited to the sialylated Fc
fragment and was
linked to an increase in expression of the inhibitory receptor FcyRIIb,
Nimmerjahn and
Ravetch., J. Exp. Med. 204:11-15 (2007).
Glycosylation in the Fc region of an antibody derived from mammalian cell
lines typically consists of a heterogeneous mix of glycoforms, with the
predominant forms
typically being comprised of the complex fucosylated glycoforms: GOF, G1F,
and, to a lesser
extent, G2F. Possible conditions resulting in incomplete galactose transfer to
the GOF
structure include, but are not limited to, non-optimized galactose transfer
machinery, such as
13-1,4 galactosyl transferase, and poor UDP-galactose transport into the Golgi
apparatus,
suboptimal cell culture and protein expression conditions, and steric
hindrance by amino acid
residues neighboring the oligosaccharide. While each of these conditions may
modulate the
ultimate degree of terminal galactose, it is thought that subsequent sialic
acid transfer to the
Fc oligosaccharide is inhibited by the closed pocket configuration of the CH2
domain. See,
for example, Fig. 1, Jefferis, R., Nature Biotech., 24 (10): 1230-1231, 2006.
Without the
correct terminal monosaccharide, specifically galactose, or with insufficient
terminal
- 24 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
galactosylated forms, there is little possibility of producing a sialylated
form, capable of
acting as a therapeutic protein, even when produced in the presence of sialyl
transferase.
Protein engineering and structural analysis of human IgG-Fc glycoforms has
shown that
glycosylation profiles are affected by Fc conformation, such as the finding
that increased
levels of galactose and sialic acid on oligosaccharides derived from CHO-
produced IgG3
could be achieved when specific amino acids from the Fc pocket were mutated,
to an alanine
including F241, F243, V264, D265 and R301. Lund et al., J. Immunol. 157(11);
4963-4969
(1996). It was further shown that certain mutations had some effect on cell
mediated
superoxide generation and complement mediated red cell lysis, which are used
as surrogate
markers for FcyRI and Clq binding, respectively.
It has been reported that yeast have been genetically engineered to produce
host strains capable of secreting glycoproteins with highly uniform
glycosylation. Choi et al.,
PNAS, USA 100(9): 5022-5027 (2003) describes the use of libraries of a 1,2
mannosidase
catalytic domains and N-acetylglucosaminyltransferase I catalytic domains in
combination
with a library of fungal type II membrane protein leader sequences to localize
the catalytic
domains to the secretory pathway. In this way, strains were isolated that
produced in vivo
glycoproteins with uniform Man5G1cNAc2 or G1cNAcMan5G1cNAc2 N-glycan
structures.
Hamilton et al., Science 313 (5792): 1441-1443 (2006) described the production
of a
glycoprotein, erythropoietin, produced in Pichia pastoris, as having a glycan
composition that
consisted predominantly of a bisialylated glycan structure, GS6.0,
NANA2Ga12G1cNAc2Man3G1cNAc2 (90.5%) and monosialylated, GS5.5,
NANAGa12G1cNAc2 Man3G1cNAc2 (7.9%). However, an antibody produced in a similar
strain will have a markedly lower content of sialylated N-glycan due to the
relatively low
level of terminal galactose substrate in the antibody as seen in Figure 4. It
has also recently
been shown that sialylation of a Fc oligosaccharide imparts anti-inflammatory
properties on
therapeutic intravenous gamma globulin and its Fc fragments, Kaneko et al.,
Science
313(5787): 670-673 (2006), and that the anti-inflammatory activity is
dependent on the a 2,6-
linked form, but not the a 2,3 form, of sialic acid, Anthony et al., Science,
320: 373-376
(2008).
- 25 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Host organisms and cell lines
The Fe-containing polypeptides of this invention can be made in any host
organism or cell line. In one embodiment, an Fe-containing polypeptide of the
invention is
made in a host cell which is capable of producing sialylated N-glycans.
In one embodiment, an Fe-containing polypeptide of the invention is made in a
mammalian cell where the cell either endogenously or through gnetic or process
manipulation
produces glycoproteins containing either a mixture of terminal a2-6 and a2-3
sialic acid, or
only terminal a2-6 sialic acid. The propagation of mammalian cells in culture
(tissue culture)
has become a routine procedure. Examples of useful mammalian host cell lines
are monkey
kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic
kidney
line (293 or 293 cells subcloned for growth in suspension culture); baby
hamster kidney cells
(BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO); mouse sertoli
cells
(TM4,); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney
cells
(VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2);
canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC
CRL
1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB
8065);
mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells; MRC 5 cells; FS4
cells;
hybridoma cell lines; NSO; SP2/0;and a human hepatoma line (Hep G2).
In one embodiment, an Fe-containing polypeptide of the invention can be
made in a plant cell which is engineered to produce sialylated N-glycans. See,
e.g., Cox et
al., Nature Biotechnology (2006) 24, 1591 - 1597 (2006) and Castilho et al.,
J. Biol. Chem.
285(21): 15923-15930 (2010).
In one embodiment, an Fe-containing polypeptide of the invention can be
made in an insect cell which is engineered to produce sialylated N-glycans.
See, e.g.,
Harrison and Jarvis, Adv. Virus Res. 68:159-91 (2006).
In one embodiment, an Fe-containing polypeptide of the invention can be
made in a bacterial cell which is engineered to produce sialylated N-glycans.
See, e.g., Lizak
et al., Bioconjugate Chem. 22:488-496 (2011).
In one embodiment, an Fe-containing polypeptide of the invention can be
made in a lower eukaryotic host cell or organism. Recent developments allow
the production
of fully humanized therapeutics in lower eukaryotic host organisms, yeast and
filamentous
fungi, such as Pichia pastoris, Gerngross et al., US Patent 7,029,872 and US
Patent No.
7,449,308, the disclosures of which are hereby incorporated by reference. See
also Jacobs et
al., Nature Protocols 4(1):58-70 (2009).
- 26 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Due to the decreased FcyR and Clq binding, the materials and methods
described herein can be used to produce recombinant glycosylated antibodies
with decreased
effector function when compared to a parent antibody. Antibodies so produced
in Pichia
pastoris by the methods of the invention were produced at high yield, with
decreased effector
function, and had a predominant species of glycoprotein having a terminal a
2,6-linked sialic
acid residue as compared to antibodies produced in glycoengineered Pichia
pastoris cells
lacking the specific Fc mutations or in Pichia pastoris host cells retaining
their endogenous
glycosylation machinery.
In one embodiment, an Fc-containing polypeptide of the invention is made in a
host cell, more preferably a yeast or filamentous fungal host cell, that has
been engineered to
produce glycoproteins having a predominant N-glycan comprising a terminal
sialic acid. In
one embodiment of the invention, the predominant N-glycan is the a 2,6 linked
form of
SA2Ga12G1cNAc2Man3G1cNAc2, produced in strains glycoengineered with a 2,6
sialyl
transferase which do not produce any a 2,3 linked sialic acid. In other
embodiments, the
strain will be engineered to express an a 2,3 sialyl transferase alone or in
combination with an
a 2,6, sialyl transferase, resulting in a 2,3 linked or a combination of a 2,6
and a 2,3 linked
sialic acid as the predominant N-glycans.
The cell lines to be used to make the Fe-containing polypeptides of the
invention can be any cell line, in particular cell lines with the capability
of producing one or
more sialylated glycoproteins. Those of ordinary skill in the art would
recognize and
appreciate that the materials and methods described herein are not limited to
the specific
strain of Pichia pastoris provided as an example herein, but could include any
Pichia pastoris
strain or other yeast or filamentous fungal strains in which N-glycans with
one or more
terminal galactose, such as Gal2G1cNAc2Man3, are produced. The terminal
galactose acts as
a substrate for the production of a 2,6-linked sialic acid, resulting in the N-
glycan structure
NANA2Ga12G1cNAc2Man3G1cNAc2. Examples of suitable strains are described in
U.S.
Pat. No. 7,029,872, US 2006-0286637 and Hamilton et al., Science 313 (5792):
1441-1443
(2006), the descriptions of which are incorporated herein as if set forth at
length.
In general, lower eukaryotes such as yeast are used for expression of the
proteins, particularly glycoproteins because they can be economically
cultured, give high
yields, and when appropriately modified are capable of suitable glycosylation.
Yeast
particularly offers established genetics allowing for rapid transformations,
tested protein
localization strategies and facile gene knock-out techniques. Suitable vectors
have expression
- 27 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
control sequences, such as promoters, including 3-phosphoglycerate kinase or
other glycolytic
enzymes, and an origin of replication, termination sequences and the like as
desired.
While the invention has been demonstrated herein using the methylotrophic
yeast Pichia pastoris, other useful lower eukaryote host cells include Pichia
pastoris, Pichia
finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens,
Pichia minuta
(Ogataea minuta, Pichia lindneri), Pichia opuntiae, Pichia thermotolerans,
Pichia salictaria,
Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia
sp.,
Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha,
Kluyveromyces sp.,
Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus
niger, Aspergillus
oryzae, Trichoderma reesei, Chrysosporiumi lucknowense, Fusarium sp., Fusarium
gramineum, Fusarium venenaturn and Neurospora crassa. Various yeasts, such as
K lactis,
Pichia pastoris, Pichia methanolica, and Hansenula polymorpha are particularly
suitable for
cell culture because they are able to grow to high cell densities and secrete
large quantities of
recombinant protein. Likewise, filamentous fungi, such as Aspergillus niger,
Fusarium sp,
Neurospora crassa and others can be used to produce glycoproteins of the
invention at an
industrial scale.
Lower eukaryotes, particularly yeast and filamentous fungi, can be genetically
modified so that they express glycoproteins in which the glycosylation pattern
is human-like
or humanized. As indicated above, the term "human-like N-glycan", as used
herein refers, to
the N-linked oligosaccharides which closely resemble the oligosaccharides
produced by non-
engineered, wild-type human cells. In preferred embodiments of the present
invention, the
host cells of the present invention are capable of producing human-like
glycoproteins with
hybrid and/or complex N-glycans; i.e., "human-like N-glycosylation." The
specific "human-
like" glycans predominantly present on glycoproteins produced from the host
cells of the
invention will depend upon the specific engineering steps that are performed.
In this manner,
glycoprotein compositions can be produced in which a specific desired
glycoform is
predominant in the composition. Such can be achieved by eliminating selected
endogenous
glycosylation enzymes and/or genetically engineering the host cells and/or
supplying
exogenous enzymes to mimic all or part of the mammalian glycosylation pathway
as
described in US Patent No. 7,449,308. If desired, additional genetic
engineering of the
glycosylation can be performed, such that the glycoprotein can be produced
with or without
core fucosylation. Use of lower eukaryotic host cells is further advantageous
in that these
cells are able to produce highly homogenous compositions of glycoprotein, such
that the
predominant glycoform of the glycoprotein may be present as greater than
thirty mole percent
-28-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
of the glycoprotein in the composition. In particular aspects, the predominant
glycoform may
be present in greater than forty mole percent, fifty mole percent, sixty mole
percent, seventy
mole percent and, most preferably, greater than eighty mole percent of the
glycoprotein
present in the composition.
Lower eukaryotes, particularly yeast, can be genetically modified so that they
express glycoproteins in which the glycosylation pattern is human-like or
humanized. Such
can be achieved by eliminating selected endogenous glycosylation enzymes
and/or supplying
exogenous enzymes as described by Gerngross et al., US Patent No. 7,449,308.
For example,
a host cell can be selected or engineered to be depleted in a1,6-mannosyl
transferase
activities, which would otherwise add mannose residues onto the N-glycan on a
glycoprotein.
In one embodiment, the host cell further includes an a1,2-mannosidase
catalytic domain fused to a cellular targeting signal peptide not normally
associated with the
catalytic domain and selected to target the al,2-mannosidase activity to the
ER or Golgi
apparatus of the host cell. Passage of a recombinant glycoprotein through the
ER or Golgi
apparatus of the host cell produces a recombinant glycoprotein comprising a
Man5G1cNAc2
glycoform, for example, a recombinant glycoprotein composition comprising
predominantly a
Man5G1cNAc2 glycoform. For example, U.S. Patent Nos. 7,029,872 and 7,449,308
and U.S.
Published Patent Application No. 2005/0170452 disclose lower eukaryote host
cells capable
of producing a glycoprotein comprising a Man5G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell further includes
a GlcNAc transferase I (GnT I) catalytic domain fused to a cellular targeting
signal peptide
not normally associated with the catalytic domain and selected to target
GleNAc transferase I
activity to the ER or Golgi apparatus of the host cell. Passage of the
recombinant
glycoprotein through the ER or Golgi apparatus of the host cell produces a
recombinant
glycoprotein comprising a GleNAcMan5G1cNAc2 glycoform, for example a
recombinant
glycoprotein composition comprising predominantly a G1eNAcMan5G1cNAc2
glycoform.
U.S. Patent Nos. 7,029,872 and 7,449,308 and U.S. Published Patent Application
No.
2005/0170452 disclose lower eukaryote host cells capable of producing a
glycoprotein
comprising a GleNAcMan5G1cNAc2 glycoform. The glycoprotein produced in the
above
cells can be treated in vitro with a hexosaminidase to produce a recombinant
glycoprotein
comprising a Man5G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell further includes
a mannosidase II catalytic domain fused to a cellular targeting signal peptide
not normally
- 29 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
associated with the catalytic domain and selected to target mannosidase II
activity to the ER
or Golgi apparatus of the host cell. Passage of the recombinant glycoprotein
through the ER
or Golgi apparatus of the host cell produces a recombinant glycoprotein
comprising a
G1cNAcMan3G1cNAc2 glycoform, for example a recombinant glycoprotein
composition
comprising predominantly a GleNAcMan3G1cNAc2 glycoform. U.S. Patent No,
7,029,872
and U.S. Published Patent Application No. 2004/0230042 discloses lower
eukaryote host
cells that express mannosidase II enzymes and are capable of producing
glycoproteins having
predominantly a G1eNAcMan3G1cNAc2 glycoform. The glycoprotein produced in the
above
cells can be treated in vitro with a hexosaminidase to produce a recombinant
glycoprotein
comprising a Man3G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell further includes
GleNAc transferase II (GnT II) catalytic domain fused to a cellular targeting
signal peptide
not normally associated with the catalytic domain and selected to target
GlcNAc transferase II
activity to the ER or Golgi apparatus of the host cell. Passage of the
recombinant
glycoprotein through the ER or Golgi apparatus of the host cell produces a
recombinant
glycoprotein comprising a G1cNAc2Man3G1cNAc2 glycoform, for example a
recombinant
glycoprotein composition comprising predominantly a GleNAc2Man3G1cNAc2
glycoform.
U.S. Patent Nos. 7,029,872 and 7,449,308 and U.S. Published Patent Application
No.
2005/0170452 disclose lower eukaryote host cells capable of producing a
glycoprotein
comprising a G1eNAc2Man3G1cNAc2 glycoform. The glycoprotein produced in the
above
cells can be treated in vitro with a hexosaminidase to produce a recombinant
glycoprotein
comprising a Man3G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell further includes
a galactosyltransferase catalytic domain fused to a cellular targeting signal
peptide not
normally associated with the catalytic domain and selected to target
galactosyltransferase
activity to the ER or Golgi apparatus of the host cell. Passage of the
recombinant
glycoprotein through the ER or Golgi apparatus of the host cell produces a
recombinant
glycoprotein comprising a GalGleNAc2 Man3G1cNAc2 or Gal2G1cNAc2Man3G1cNAc2
glycoform, or mixture thereof for example a recombinant glycoprotein
composition
comprising predominantly a GalGleNAc2 Man3G1cNAc2 glycoform or
Gal2G1cNAc2Man3G1cNAc2 glycoform or mixture thereof. U.S. Patent No, 7,029,872
and
U.S. Published Patent Application No. 2006/0040353 discloses lower eukaryote
host cells
- 30 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
capable of producing a glycoprotein comprising a Ga12G1cNAc2 Man3G1cNAc2
glycoform.
The glycoprotein produced in the above cells can be treated in vitro with a
galactosidase to
produce a recombinant glycoprotein comprising a GlcNAc2Man3 G1cNAc2 glycoform,
for
example a recombinant glycoprotein composition comprising predominantly a
G1cNAc2Man3G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell further includes
a sialyltransferase catalytic domain fused to a cellular targeting signal
peptide not normally
associated with the catalytic domain and selected to target sialyltransferase
activity to the ER
or Golgi apparatus of the host cell. In a preferred embodiment, the
sialyltransferase is an
alpha2,6-sialyltransferase. Passage of the recombinant glycoprotein through
the ER or Golgi
apparatus of the host cell produces a recombinant glycoprotein comprising
predominantly a
NANA2Ga12G1cNAc2Man3G1cNAc2 glycoform or NANAGa12G1cNAc2Man3G1cNAc2
glycoform or mixture thereof. For lower eukaryote host cells such as yeast and
filamentous
fungi, it is useful that the host cell further include a means for providing
CMP-sialic acid for
transfer to the N-glycan. U.S. Published Patent Application No. 2005/0260729
discloses a
method for genetically engineering lower eukaryotes to have a CMP-sialic acid
synthesis
pathway and U.S. Published Patent Application No. 2006/0286637 discloses a
method for
genetically engineering lower eukaryotes to produce sialylated glycoproteins.
To enhance the
amount of sialylation, it can be advantageous to construct the host cell to
include two or more
copies of the CMP-sialic acid synthesis pathway or two or more copies of the
sialylatransferase. The glycoprotein produced in the above cells can be
treated in vitro with a
neuraminidase to produce a recombinant glycoprotein comprising predominantly a
Ga12G1cNAc2Man3G1cNAc2 glycoform or GalGleNAc2Man3G1cNAc2 glycoform or
mixture thereof.
Any one of the preceding host cells can further include one or more GlcNAc
transferase selected from the group consisting of GnT III, GnT IV, GnT V, GnT
VI, and GnT
IX to produce glycoproteins having bisected (GnT III) and/or multiantennary
(GnT IV, V, VI,
and IX) N-glycan structures such as disclosed in U.S. Published Patent
Application Nos.
2005/0208617 and 2007/0037248. Further, the proceeding host cells can produce
recombinant glycoproteins (for example, antibodies) comprising SA(1-4)Gal(1-
4)G1cNAc(2-
Man3G1cNAc2, including antibodies comprising NANA (1-4)Gal(1-4)G1cNAc(2-4)
Man3G1cNAc2, NGNA(1-4)Gal(1-4)G1cNAc(2-4)Man3G1cNAc2 or a combination of
NANA (1-4)Gal(1-4)G1cNAc(2-4) Man3G1cNAc2 and NGNA(1-4)Gal(1-4)G1cNAc(2-4)
-31 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Man3G1cNAc2. In one embodiment, the recombinant glycoprotein will comprise N-
glycans
comprising a structure selected from the group consisting of SA(1-4)Gal(1-
4)G1cNAc(2-4)
Man3G1cNAc2 and devoid of any a2-3 linked SA.
In further embodiments, the host cell that produces glycoproteins that have
predominantly G1eNAcMan5G1cNAc2 N-glycans further includes a
galactosyltransferase
catalytic domain fused to a cellular targeting signal peptide not normally
associated with the
catalytic domain and selected to target the galactosyltransferase activity to
the ER or Golgi
apparatus of the host cell. Passage of the recombinant glycoprotein through
the ER or Golgi
apparatus of the host cell produces a recombinant glycoprotein comprising
predominantly the
Ga1G1eNAcMan5G1cNAc2 glycoform.
In a further embodiment, the immediately preceding host cell that produced
glycoproteins that have predominantly the GalGleNAcMan5G1cNAc2 N-glycans
further
includes a sialyltransferase catalytic domain fused to a cellular targeting
signal peptide not
normally associated with the catalytic domain and selected to target
sialyltransferase activity
to the ER or Golgi apparatus of the host cell. Passage of the recombinant
glycoprotein
through the ER or Golgi apparatus of the host cell produces a recombinant
glycoprotein
comprising a SAGa1G1cNAcMan5G1eNAc2 glycoform (for example
NANAGa1G1eNAcMan5G1cNAc2 or NGNAGa1G1eNAcMan5G1cNAc2 or a mixture
thereof).
Any of the preceding host cells can further include one or more sugar
transporters such as UDP-GleNAc transporters (for example, Kluyveromyces
lactis and Mus
muscu/us UDP-G1cNAc transporters), UDP-galactose transporters (for example,
Drosophila
melanogaster UDP-galactose transporter), and CMP-sialic acid transporter (for
example,
human sialic acid transporter). Because lower eukaryote host cells such as
yeast and
filamentous fungi lack the above transporters, it is preferable that lower
eukaryote host cells
such as yeast and filamentous fungi be genetically engineered to include the
above
transporters.
Further, any of the preceding host cells can be further manipulated to
increase
N-glycan occupancy. See e, g., Gaulitzek et al., Biotechnol. Bioengin.
103:1164-1175
(2009); Jones et al., Biochim. Biospyhs. Acta 1726:121-137 (2005);
W02006/107990. In
one embodiment, any of the preceding host cells can be further engineered to
comprise at
least one nucleic acid molecule encoding a heterologous single-subunit
oligosaccharyltransferase (for example, Leishmania sp. STT3A protein, STT3B
protein,
- 32 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
STT3C protein, STT3D protein or combinations thereof) and a nucleic acid
molecule
encoding the heterologous glycoprotein, and wherein the host cell expresses
the endogenous
host cell genes encoding the proteins comprising the endogenous OTase complex.
In one
embodiment, any of the preceding host cells can be further engineered to
comprise at least
one nucleic acid molecule encoding a Leishmania sp. STT3D protein and a
nucleic acid
molecule encoding the heterologous glycoprotein, and wherein the host cell
expresses the
endogenous host cell genes encoding the proteins comprising the endogenous
OTase
complex.
Host cells further include lower eukaryote cells (e.g., yeast such as Pichia
pastoris) that are genetically engineered to produce glycoproteins that do not
have a-
mannosidase-resistant N-glycans. This can be achieved by deleting or
disrupting one or more
of the 13-mannosyltransferase genes (e.g., BMT1, BMT2, BMT3, and BMT4) (See,
U.S.
Published Patent Application No. 2006/0211085) and glycoproteins having
phosphomannose
residues by deleting or disrupting one or both of the phosphomannosyl
transferase genes
PNO1 and MNN4B (See for example, U.S. Patent Nos. 7,198,921 and 7,259,007),
which in
further aspects can also include deleting or disrupting the MNN4A gene.
Disruption includes
disrupting the open reading frame encoding the particular enzymes or
disrupting expression
of the open reading frame or abrogating translation of RNAs encoding one or
more of the 13-
mannosyltransferases and/or phosphomannosyltransferases using interfering RNA,
antisense
RNA, or the like. Further, cells can produce glycoproteins with a-mannosidase-
resistant N-
glycans through the addition of chemical hinhibios or through modification of
the cell culture
condition. These host cells can be further modified as described above to
produce particular
N-glycan structures.
Host cells further include lower eukaryote cells (e.g., yeast such as Pichia
pastoris) that are genetically modified to control 0-glycosylation of the
glycoprotein by
deleting or disrupting one or more of the protein 0-mannosyltransferase (Dol-P-
Man:Protein
(Ser/Thr) Mannosyl Transferase genes) (PMI's) (See U.S. Patent No. 5,714,377)
or grown in
the presence of Pmtp inhibitors and/or an a -mannosidase as disclosed in
Published
International Application No. WO 2007/061631, or both. Disruption includes
disrupting the
open reading frame encoding the Pmtp or disrupting expression of the open
reading frame or
abrogating translation of RNAs encoding one or more of the Pmtps using
interfering RNA,
antisense RNA, or the like. The host cells can further include any one of the
aforementioned
host cells modified to produce particular N-glycan structures.
- 33 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Pmtp inhibitors include but are not limited to a benzylidene
thiazolidinediones. Examples of benzylidene thiazolidinediones that can be
used are 5-[[3,4-
bis(phenylmethoxy) phenyl]methylene]-4-oxo-2-thioxo-3-thiazolidineacetic Acid;
5-[[3-(1-
Phenylethoxy)-4-(2-phenylethoxy)]phenyl]methylene]-4-oxo-2-thioxo-3-
thiazolidineacetic
Acid; and 5-[[3-(1-Pheny1-2-hydroxy)ethoxy)-4-(2-
phenylethoxy)]phenyllmethylene]-4-oxo-
2-thioxo-3-thiazolidineacetic acid.
In particular embodiments, the function or expression of at least one
endogenous PMT gene is reduced, disrupted, or deleted. For example, in
particular
embodiments the function or expression of at least one endogenous PMT gene
selected from
the group consisting of the PMT1, PMT2, PMT3, and PMT4 genes is reduced,
disrupted, or
deleted; or the host cells are cultivated in the presence of one or more PMT
inhibitors. In
further embodiments, the host cells include one or more PMT gene deletions or
disruptions
and the host cells are cultivated in the presence of one or more Pmtp
inhibitors. In particular
aspects of these embodiments, the host cells also express a secreted a-1,2-
mannosidase.
PMT deletions or disruptions and/or Pmtp inhibitors control 0-glycosylation
by reducing 0-glycosylation occupancy, that is, by reducing the total number
of 0-
glycosylation sites on the glycoprotein that are glycosylated. The further
addition of an a -
1,2-mannosidase that is secreted by the cell controls 0-glycosylation by
reducing the mannose
chain length of the 0-glycans that are on the glycoprotein. Thus, combining
PMT deletions
or disruptions and/or Pmtp inhibitors with expression of a secreted a-1,2-
mannosidase
controls 0-glycosylation by reducing occupancy and chain length. In particular
circumstances, the particular combination of PMT deletions or disruptions,
Pmtp inhibitors,
and a-1,2-mannosidase is determined empirically as particular heterologous
glycoproteins
(Fabs and antibodies, for example) may be expressed and transported through
the Golgi
apparatus with different degrees of efficiency and thus may require a
particular combination
of PMT deletions or disruptions, Pmtp inhibitors, and a-1,2-mannosidase. In
another aspect,
genes encoding one or more endogenous mannosyltransferase enzymes are deleted.
This
deletion(s) can be in combination with providing the secreted a-1,2-
mannosidase and/or PMT
inhibitors or can be in lieu of providing the secreted a-1,2-mannosidase
and/or PMT
inhibitors.
Thus, the control of 0-glycosylation can be useful for producing particular
glycoproteins in the host cells disclosed herein in better total yield or in
yield of properly
assembled glycoprotein. The reduction or elimination of 0-glycosylation
appears to have a
beneficial effect on the assembly and transport of whole antibodies and Fab
fragments as they
- 34 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
traverse the secretory pathway and are transported to the cell surface. Thus,
in cells in which
0-glycosylation is controlled, the yield of properly assembled antibodies or
Fab fragments is
increased over the yield obtained in host cells in which 0-glycosylation is
not controlled.
To reduce or eliminate the likelihood of N-glycans and 0-glycans with 0-
linked mannose residues, which are resistant to a-mannosidases, the
recombinant
glycoengineered Pichia pastoris host cells are genetically engineered to
eliminate
glycoproteins having a-mannosidase-resistant N-glycans by deleting or
disrupting one or
more of the P-mannosyltransferase genes (e.g., BMT1, BMT2, BMT3, and BMT4)
(See, U.S.
Patent No. 7,465,577 and U.S. Patent No. 7,713,719). The deletion or
disruption of BMT2
and one or more of BMT1, BMT3, and BMT4 also reduces or eliminates detectable
cross
reactivity to antibodies against host cell protein.
Yield of glycoprotein can in some situations be improved by overexpressing
nucleic acid molecules encoding mammalian or human chaperone proteins or
replacing the
genes encoding one or more endogenous chaperone proteins with nucleic acid
molecules
encoding one or more mammalian or human chaperone proteins. In addition, the
expression
of mammalian or human chaperone proteins in the host cell also appears to
control 0-
glycosylation in the cell. Thus, further included are the host cells herein
wherein the function
of at least one endogenous gene encoding a chaperone protein has been reduced
or
eliminated, and a vector encoding at least one mammalian or human homolog of
the
chaperone protein is expressed in the host cell. Also included are host cells
in which the
endogenous host cell chaperones and the mammalian or human chaperone proteins
are
expressed. In further aspects, the lower eukaryotic host cell is a yeast or
filamentous fungi
host cell. Examples of the use of chaperones of host cells in which human
chaperone proteins
are introduced to improve the yield and reduce or control 0-glycosylation of
recombinant
proteins has been disclosed in Published International Application No. WO
2009105357 and
W02010019487 (the disclosures of which are incorporated herein by reference).
Like above,
further included are lower eukaryotic host cells wherein, in addition to
replacing the genes
encoding one or more of the endogenous chaperone proteins with nucleic acid
molecules
encoding one or more mammalian or human chaperone proteins or overexpressing
one or
more mammalian or human chaperone proteins as described above, the function or
expression
of at least one endogenous gene encoding a protein 0-mannosyltransferase (PMT)
protein is
reduced, disrupted, or deleted. In particular embodiments, the function of at
least one
endogenous PMT gene selected from the group consisting of the PMT], PMT2,
PMT3, and
PMT4 genes is reduced, disrupted, or deleted.
- 35 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In addition, 0-glycosylation may have an effect on an antibody or Fab
fragment's affinity and/or avidity for an antigen. This can be particularly
significant when the
ultimate host cell for production of the antibody or Fab is not the same as
the host cell that
was used for selecting the antibody. For example, 0-glycosylation might
interfere with an
antibody's or Fab fragment's affinity for an antigen, thus an antibody or Fab
fragment that
might otherwise have high affinity for an antigen might not be identified
because 0-
glycosylation may interfere with the ability of the antibody or Fab fragment
to bind the
antigen. In other cases, an antibody or Fab fragment that has high avidity for
an antigen
might not be identified because 0-glycosylation interferes with the antibody's
or Fab
fragment's avidity for the antigen. In the preceding two cases, an antibody or
Fab fragment
that might be particularly effective when produced in a mammalian cell line
might not be
identified because the host cells for identifying and selecting the antibody
or Fab fragment
was of another cell type, for example, a yeast or fungal cell (e.g., a Pichia
pastoris host cell).
It is well known that 0-glycosylation in yeast can be significantly different
from 0-
glycosylation in mammalian cells. This is particularly relevant when comparing
wild type
yeast 0-glycosylation with mucin-type or dystroglycan type 0-glycosylation in
mammals. In
particular cases, 0-glycosylation might enhance the antibody or Fab fragments
affinity or
avidity for an antigen instead of interfere with antigen binding. This effect
is undesirable
when the production host cell is to be different from the host cell used to
identify and select
the antibody or Fab fragment (for example, identification and selection is
done in yeast and
the production host is a mammalian cell) because in the production host the 0-
glycosylation
will no longer be of the type that caused the enhanced affinity or avidity for
the antigen.
Therefore, controlling 0-glycosylation can enable use of the materials and
methods herein to
identify and select antibodies or Fab fragments with specificity for a
particular antigen based
upon affinity or avidity of the antibody or Fab fragment for the antigen
without identification
and selection of the antibody or Fab fragment being influenced by the 0-
glycosylation system
of the host cell. Thus, controlling 0-glycosylation further enhances the
usefulness of yeast or
fungal host cells to identify and select antibodies or Fab fragments that will
ultimately be
produced in a mammalian cell line.
Those of ordinary skill in the art would further appreciate and understand how
to utilize the methods and materials described herein in combination with
other Pichia
pastoris and yeast cell lines that have been genetically engineered to produce
specific N-
glycans or sialylated glycoproteins, such as, but, not limited to, the host
organisms and cell
lines described above that have been genetically engineered to produce
specific galactosylated
-36-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
or sialylated forms. See, for example, US 2006-0286637, Production of
Sialylated N-Glycans
in Lower Eukaryotes, in which the pathway for galactose uptake and utilization
as a carbon
source has been genetically modified, the description of which is incorporated
herein as if set
forth at length.
Additionally, the methods herein can be used to produce the above described
recombinant Fc-containing polypeptides in other lower eukaryotic cell lines
which have been
engineered to produce human-like and human glycoproteins that do not have a2,6
sialyltransferase activity. The methods can also be used to produce the above
described
recombinant Fc-containing polypeptides in eukaryotic cell lines in which
production of
sialylated N-glycans is an innate feature.
Levels of a2,3-and a2,6-linked sialic acid on the Fc-containing polypeptides
can be measured using well known technicques including nuclear magnetic
resonance
(NMR), normal phase high performance liquid chromatography (HPLC), and high
performance anion exchange chromatography with pulsed amperometric detection
(HPAEC-
PAD).
Biological Properties of Fc muteins
For many Fc-containing polypeptides the lack of or significant decrease in
effector function, as shown by decreased FcyR and Clq binding, Idusogie et
al., J.
Immunology, 164(8): 4178-84 (2000) and Shields et al., J. Biol. Chem., 276:
6591-6604
(2001), and increased anti-inflammatory properties would be desirable
characteristics.
Applicants have developed a quadruple Fc mutein,
F243A/V264A/S267E/L328F, which will produce Fc-containing polypeptides having
the
aforesaid desired characteristics. The Examples herein comprise transforming a
host cell with
a polynucleotide vector encoding a Fc-containing polypeptide comprising
mutations at
positions 243, 264, 267 and 328 of the Fc region, and culturing the
transformed host cell to
produce the Fc-containing polypeptide.
Production of Fc-containing polypeptides
The Fc-containing polypeptides of the invention can be made according to any
method known in the art suitable for generating polypeptides comprising an Fc
region. In one
embodiment, the Fc-containing polypeptide is an antibody or an antibody
fragment
(including, without limitation a polypeptide comprising, consisting, or
consisting essentially
of the Fc region of an antibody). In another embodiment, the Fc-containing
polypeptide is an
- 37 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
immunoadhesin. Methods of preparing antibody and antibody fragments are well
known in
the art. Methods of introducing point mutations into a polypeptide, for
example site directed
mutagenesis, are also well known in the art.
In the Examples disclosed herein, an IgG1 heavy and light chain containing a
consensus C112 sequence and the Fc double mutants described herein were
expressed in two
different glycoengineered Pichia pastoris strains. As described in the
Examples that follow,
the heavy and light chain gene sequences were under the control of a methanol
inducible
promoter, A0X1, and incorporated a bleomycin (Zeocin) selection marker. This
strategy
integrates the entire expression cassette into the Trp2 locus by homologous
DNA
recombination.
Secreted antibodies were captured from the fermentation broth by protein A
affinity chromatography followed by a Source 30S cation exchange purification
step.
Purified antibodies were characterized by SDS-PAGE (Figure 2) to assess proper
assembly.
As seen in Figure 2, the antibodies produced by the materials and methods
herein were
properly assembled.
Antigen affinity for the various antibodies made by the materials and methods
herein was determined by a cell based assay using a Biacore. As expected, all
of the
antibodies, including the Fc muteins, bound equally well to the PCSK9 antigen.
N-Glycan analysis of Fc muteins
For many glycoproteins, including certain antibodies, sialylation of the
terminal N-linked glycan of an IgG Fc region is essential for producing
glycoproteins and
antibodies that have the correct conformation to impart therapeutic activity.
See, for
example, Anthony et al., Science, 320: 373-376 (2008), where terminal
sialylation was
correlated to anti-inflammatory activity for an IVIG preparation. Sialylation
requires the
presence of a penultimate galactose, upon which the sialyl transferase acts to
form the
sialylated glycan. Thus, glycoproteins lacking one or more terminal galactose
glycoforms
cannot produce antibodies having the a2,6-linked sialic acid composition
associated with
anti-inflammatory activity.
Mammalian cells have full capability of sialylation on its glycoproteins,
however, due to spatial constricts antibodies produced in mammalian cell
culture, such as
CHO cells, have even incomplete galactose transfer to its N297 linked glycans.
Moreover,
because of further spatial hindrance, the level of sialylation of antibody
from mammalian cell
such as CHO usually contains little or no sialic acid at its N297 linked
glycans. In the case of
-38-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
CHO cell production, when sialic acid is added, it is linked by an a2,3-
linkage. CHO cells do
not express an a2,6 sialyl transferase necessary to produce the a2,6-linked
form of sialic acid,
which has been associated with anti-inflammatory activity (Lee et al, J. Biol.
Chem. 264:
13848-13855 (1989). Overexpression of a specific a2,6 sialyltranferase in CHO
can give rise
to a mixture of a2,3-linked and a2,6-linked sialic acid (Bragonzi et al., BBA
1474:273-282
(2000); Biochem. Biophys. Res. Comm. 289: 243-249 (2001)).
Glycoengineered Pichia pastoris GFI5.0 strains, which are capable of
producing high levels of galactosylated non-antibody proteins, such as
erythropoietin
(Hamilton et al., Science, 313: 1441-1443 (2006)), produce antibodies with
relatively low
amounts of a terminal galactose that can be acted upon to form the a 2,6-
linked sialylated
form. Antibodies produced in such Pichia pastoris strains typically have a
composition
including glycoforms GO (60%), G1 (17%), G2 (4%) and Man5 (8%). Even
antibodies
produced in Pichia pastoris GFI6.0 strains, which have a glycan composition
comprising GO
(43.5%), G1 (20.8%), G2(2.7%), NANAGa1G1eNAcMan5G1cNAc2 (5.5%), and
NANAGa12G1cNAc2Man3G1cNAc2 (4.9%), have relatively low levels of the a 2,6-
linked
sialylated form. Thus, antibodies produced in GFI 5.0 and 6.0 strains have
much lower levels
of galactosylation and sialylation compared to non-antibody proteins (such as
erythropoietin)
produced in the same strains.
Glycoforms of the Pichia pastoris wild-type PCSK-9 antibody (1F11) from the
GFI5.0 strain (YGLY21351) included M4, GO, Gl, M6, G2 glycans. Glycoforms of
the
Pichia pastoris double mutein PCSK-9 antibody (1F11 F243A/V264A) from the GFI
6.0
strain (YGLY23258) included GO, M5, Gl, G2, Al and A2 glycans. Glycoforms of
the
Pichia pastoris 1F11 F243A/V264A/S267E/L328F) from the GFI 6.0 strain
included: Al,
Al H and A2 (YGLY25267) and G2, Man5, Al and A2 (YGLY25269).
- 39 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
FcyR binding of Fe muteins
Using an ELISA based assay, Applicants compared Fc gamma receptor (Fc7R)
binding for the commercially available Herceptin, a wildtype anti-PCSK9
antibody (1F11), an
double mutein anti-PCSK9 (1F11) antibody having the F243AN243A mutations, and
a
quadruple mutein anti-PCSK9 (1F11) antibody having the F243AN264A/S267E/L328F
mutations. As shown in the Examples, the Fe quadruple mutein has decreased
affinity to
FcyRI, Fc7RlIa and FeyRIIa, and increased affinity to Fc7RIIb, as compared to
an antibody
having a native Fe region or as compared to an antibody having a F243A/V264A
double
mutation.
Neither the double nor the quadruple mutein bind Fe7RIIa.
Neither the double nor the quadruple mutein bind Fc7RIIIa-F158 or FcyRIIIa-
V158.
However, the quadruple mutein has increased binding to Fc71ZIIb which is
surprising since the duble mutein shown only minimal binding to this receptor.
Taken together, these data suggest that the quadruple Fe mutein is less prone
to activating and recruiting immune cells such as macrophages, monocytes and
natural killer
cells as compared to double mutein or non-mutein (native) versions of the
antibody.
Biological Targets
It should be noted that while, in the examples that follow, Applicants
exemplify the materials and methods of the invention using an anti-PCSK9
antibody, the
invention is not limited to the disclosed antibodies. Those of ordinary skill
in the art would
recognize and appreciate that the materials and methods herein could be used
to produce any
Fe-containing polypeptide for which the characteristics of enhanced anti-
inflammatory
activity or decreased effector function would be desirable. It should further
be noted that
there is no restriction as to the type of Fe-containing polypeptide or
antibody so produced by
the invention. The Fe region of the Fe-containing polypeptide could be from an
IgA, IgD,
IgE, IgG or IgM. In one embodiment, the Fe region of the Fe-containing
polypeptide is from
an IgG, including IgGl, IgG2, IgG3 or IgG4. In one embodiment, Fe region of
the Fe-
containing polypeptide is from an IgG 1 . In specific embodiments the
antibodies or antibody
fragments produced by the materials and methods herein can be humanized,
chimeric or
human antibodies.
In some embodiments, the Fe-containing polypeptides of the invention will
bind to a biological target that is involved in inflammation.
- 40 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In some embodiments, the Fe-containing polypeptide of the invention will
bind to a pro-inflammatory cytokine. In some embodiments, the Fe-containing
polypeptide of
the invention will bind to a molecule selected from the group consisting of:
APRIL, INF- a,
BAFF (BLys), CD22, TNF-a, IL-1, IL-2, IL-4, IL-5, IL-6, IL-8, IL-9, IL-10, IL-
12, IL-15, IL-
17, IL-18, IL-20, IL-21, IL-22, IL-23, IL-23R, IL-25, IL-27, IL-33, CD2, CD4,
CD11A,
CD14, CD18, CD19, CD23, CD25, CD38, CD40, CD4OL, CD20, CD52, CD64, CD80,
CD147, CD200, CD200R, TSLP, TSLPR, PD-1, PDL1, CTLA4, VLA-4, VEGF, PCSK9,
a47-integrin, E-selectin, Fact II, ICAM-3, beta2-integrin, IFNy, C5, CBL,
LCAT, CR3,
MDL-1, GITR, ADDL, CGRP, TRKA, IGF1R, RANKL, GTC, aBLys, or the receptor for
any
of the above mentioned molecules. In one embodiment, the Fe-containing
polypeptide of the
invention will bind to TNF-a. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to Her2. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to PCSK9. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to TNFR. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to LCAT. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to TSLP. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to PD-1. In another embodiment, the Fe-containing
polypeptide of the
invention will bind to IL-23.
In some embodiments, the Fe-containing polypeptides of the invention will be
specific for an antigen selected from autoimmune antigens, allergens, MHC
molecules or
Rhesus factor D antigen. See, e.g., the antigens listed in Table 1 of
W02010/10910, which
is incorporated herein by reference.
Methods of Increasing Anti-Inflammatory Properties or Decreasing Effector
Function/Cytotoxicity
The invention also comprises a method of increasing the anti-inflammatory
properties of an Fe-containing polypeptide comprising: selecting a parent Fe-
containing
polypeptide that is useful in treating an inflammatory condition (for example,
an antibody or
immunoadhesin that binds to an antigen that is involved in inflammation) and
introducing
mutations at positions 243, 264, 267 and 328 of the Fe-containing polypeptide,
wherein the
numbering is according to the EU index as in Kabat, wherein the Fe-containing
polypeptide
has increased anti-inflammatory properties when compared to the parent Fe-
containing
polypeptide. In one embodiment, the Fe-containing polypeptide comprises
mutations F243A,
V264A, S267E, and L328F. In one embodiment, the parent Fe-containing
polypeptide is an
-41-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
antibody, antibody fragment or immunoadhesin that binds to an antigen that is
involved in
inflammation. In one embodiment, the parent Fc-containing polypeptide is an an
antibody,
antibody fragment or immunoadhesin that is already marketed or under
development for the
treatment of an inflammatory conditions. In another embodiment, the parent Fc-
contanining
polypeptide is an antibody selected from the group consisting of: Muromonab-
CD3 (anti-CD3
receptor antibody), Abciximab (anti-CD41 7E3 antibody), Rituximab (anti-CD20
antibody),
Daclizumab (anti-CD25 antibody), Basiliximab (anti-CD25 antibody), Palivizumab
(anti-
RSV (respiratory syncytial virus) antibody), Infliximab (anti-TNFa antibody),
Trastuzumab
(anti-Her2 antibody), Gemtuzumab ozogamicin (anti-CD33 antibody), Alemtuzumab
(anti-
CD52 antibody), Ibritumomab tiuxeten (anti-CD20 antibody), Adalimumab (anti-
TNFa
antibody), Omalizumab (anti-IgE antibody), Tositumomab-131I (iodinated
derivative of an
anti-CD20 antibody), Efalizumab (anti-CD11 a antibody), Cetuximab (anti-EGF
receptor
antibody), Golimumab (anti-TNFa antibody), Bevacizumab (anti VEGF-A antibody),
Natalizumab (anti a4 integrin), Efalizumab (anti CD11 a), Cetolizumab (anti-
TNFa antibody)
, Tocilizumab (anti-IL-6R), Ustenkinumab (anti IL-12/23), alemtuzumab (anti
CD52), and
natalizumab (anti a4 integrin), and variants thereof. In another embodiment,
the parent Fc-
contanining polypeptide is an Fe-fusion protein selected from the group
consisting of:
Arcalyst/ rilonacept (IL1R-Fc fusion), Oreneia/ abatacept (CTLA-4-Fc fusion),
Amevive/
alefacept (LFA-3-Fc fusion), Anakinra-Fc fusion (IL-1Ra-Fc fusion protein),
etanercept
(TNFR-Fc fusion protein), FGF-21-Fc fusion protein, GLP-1-Fc fusion protein,
RAGE-Fc
fusion protein, ActRIIA-Fc fusion protein, ActRIIB-Fe fusion protein, glucagon-
Fc fusion
protein, oxyntomodulin-Fc-fusion protein, GM-CSF-Fc fusion protein, EPO-Fc
fusion
protein, Insulin-Fe fusion protein, proinsulin-Fc fusion protein and insulin
precursor-Fe
fusion protein, and analogs and variants thereof.
The invention also comprises a method of reducing the effector function of an
Fe-containing polypeptide, comprising introducing mutations at positions 243,
264, 267 and
328 of a parent Fc-contaning polypeptide, wherein said Fe containing
polypeptide has
decreased effector function when compared to the parent Fe-containing
polypeptide, wherein
the numbering is according to the EU index as in Kabat. In one embodiment, the
Fe-
containing polypeptide comprises mutations F243A, V264A, S267E, and L328F. In
one
embodiment, the Fe-containing polypeptide is an antibody or antigen binding
fragment
thereof In one embodiment, the effector function is ADCC. In another
embodiment, the
effector function is CDC.
- 42 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
The invention also comprises a method of decreasing cytotoxicity of an Fe-
containing polypeptide comprising: selecting a parent Fc-containing
polypeptide that is useful
in treating an inflammatory condition (for example, an antibody or
immunoadhesin that binds
to an antigen that is involved in inflammation) that binds to an antigen that
is involved in
inflammation and introducing mutations at positions 243, 264, 267 and 328 of
the Fe-
containing polypeptide, wherein the numbering is according to the EU index as
in Kabat,
wherein the Fc-containing polypeptide has decreased cytotoxicity when compared
to the
parent Fc-containing polypeptide. In a embodiment, the Fc-containing
polypeptide comprises
mutations F243A, V264A, S267E, and L328F.
In one embodiment, the parent Fc-containing polypeptide comprises a native
Fc region. In another embodiment, the parent Fc-containing polypeptide
comprises a F243A
mutation. In another embodiment, the parent Fc-containing polypeptide
comprises a V264A
mutation. In another embodiment, the parent Fc-containing polypeptide
comprises a
F243A/V264A mutation.
Methods of Treatment
The invention also comprises a method of treating an inflammatory condition
in a subject in need thereof comprising: administering to the subject a
therapeutically
effective amount of an Fc-containing polypeptide comprising mutations at
positions 243, 264,
267 and 328, wherein the numbering is according to the EU index as in Kabat.
In one
embodiment, the Fc-containing polypeptide comprises mutations F243A, V264A,
S267E, and
L328F. In one embodiment, the Fc-containing polypeptide is an antibody
fragment
comprising SEQ ID NO:7. In another embodiment, the Fc-containing polypeptide
is an
antibody fragment comprising SEQ ID NO:8. In another embodiment, the Fc-
containing
polypeptide of the invention is an antibody fragment comprising SEQ ID NO:17.
In another
embodiment, the Fc-containing polypeptide is an antibody fragment consisting
(or consisting
essentially of) SEQ ID NO:7 or SEQ ID NO:8 or SEQ ID NO:17. The Fc-containing
polypeptide of the invention can be administered by any route. In one
embodiment, the Fc-
containing polypeptide is administered parenterally. In one embodiment, the Fc-
containing
polypeptide is administered subcutaneously.
In one embodiment, the inflammatory condition is unwanted inflammatory
immune reactions.
In one embodiment, the inflammatory condition is an autoimmune disease. In
one embodiment, the inflammatory condition will be multiple sclerosis. In one
embodiment,
-43 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
the inflammatory condition is systemic lupus erythematosus. In one embodiment,
the
inflammatory condition is type I diabetes.
In one embodiment, the inflammatory condition is a primary
immunodeficiency syndrome, including congential agammaglobulinaemia and
hypogammaglobulinaemia, common variable immunodeficiency, severed combined
immunodeficiency, or Wiskott Aldrich syndrome.
In one embodiment, the inflammatory condition is a secondary
immunodeficiency syndrome, including B-cell lymphocytic leukemia, HIV
infection or an
allogeneic bone marrow transplantation.
In one embodiment, the inflammatory condition is idiopathic
thrombocytopenic purpura.
In one embodiment, the inflammatory condition is multiple myeloma.
In one embodiment, the inflammatory condition is Guillain-Barre syndrome.
In one embodiment, the inflammatory condition is Kawasaki disease.
In one embodiment, the inflammatory condition is chronic inflammatory
demyelinating polyneropathy (CIDP).
In one embodiment, the inflammatory condition is autoimmune nuetropenia.
In one embodiment, the inflammatory condition is hemolytic anemia.
In one embodiment, the inflammatory condition is anti-Factor VIII
autoimmune disease.
In one embodiment, the inflammatory condition is multifocal neuropathy.
In one embodiment, the inflammatory condition is systemic vasculitis (ANCA
positive).
In one embodiment, the inflammatory condition is polymyositis.
In one embodiment, the inflammatory condition is dermatomyositis.
In one embodiment, the inflammatory condition is antiphospholipid syndrome.
In one embodiment, the inflammatory condition is sepsis syndrome.
In one embodiment, the inflammatory condition is graft-v-host disease.
In one embodiment, the inflammatory condition is allergy.
In one embodiment, the inflammatory condition is an anti-Rhesus factor D
reaction.
In one embodiment, the inflammatory condition is systemic lupus
eythematusus (SLU).
- 44 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
In one embodiment, the inflammatory condition is an inflammatory condition
of the cardiovascular system. The Fc-containing polypeptides of the invention
may be used to
treat atherosclerosis, atherothrombosis, coronary artery hypertension, acute
coronary
syndrome and heart failure, all of which are associated with inflammation.
In one embodiment, the inflammatory condition is an inflammatory condition
of the central nervous system. In another embodiment, the inflammatory
condition will be an
inflammatory condition of the peripheral nervous system. For example, the Fc-
containing
polypeptides of the invention may be used for the treatment of, e.g.,
Alzheimer's disease,
amyotrophic lateral sclerosis (a.k.a. ALS; Lou Gehrig's disease), ischemic
brain injury, prion
diseases, and HIV-associated dementia.
In one embodiment, the inflammatory condition is an inflammatory condition
of the gastrointestinal tract. For example, the Fc-containing polypeptides of
the invention
may be used for treating inflammatory bowel disorders, e.g., Crohn's disease,
ulcerative
colitis, celiac disease, and irritable bowel syndrome.
In one embodiment, the inflammatory condition is psoriasis, atopic dermatitis,
arthritis, including rheumatoid arthritis, osteoarthritis, and psoriatic
arthritis.
In one embodiment, the inflammatory condition is steroid-dependent atopic
dermatitis.
In one embodiment, the inflammatory condition is cachexia.
Examples of other inflammatory disorders that can be treated using the Fc-
containing polypeptides of the invention also include: acne vulgaris, asthma,
autoimmune
diseases, chronic prostatitis, glomerulonephritis, hypersensitivities, pelvic
inflammatory
disease, reperfusion injury, sarcoidosis, transplant rejection, vasculitis,
interstitial cystitis and
myopathies.
In one embodiment, the Fc-containing polypeptide of the invention will be
administered a dose of between 1 to 100 milligrams per kilograms of body
weight. In one
embodiment, the Fc-containing polypeptide of the invention will be
administered a dose of
between 0.001 to 10 milligrams per kilograms of body weight. In one
embodiment, the Fc-
containing polypeptide of the invention will be administered a dose of between
0.001 to 0.1
milligrams per kilograms of body weight. In one embodiment, the Fc-containing
polypeptide
of the invention will be administered a dose of between 0.001 to 0.01
milligrams per
kilograms of body weight.
Pharmaceutical Formulations
- 45 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
The invention also comprises pharmaceutical formulations comprising an Fe-
containing polypeptide of the invention and a pharmaceutically acceptable
carrier.
In one embodiment, the invention relates a pharmaceutical composition
comprising an Fc-containing polypeptide, wherein at least 70% of the N-glycans
on the Fc-
containing polypeptide comprise an oligosaccharide structure selected from the
group
consisting of NANA(1_4)Gal(1_4)G1cNAc(2_4)Man3G1cNAcz wherein the Fc-
containing
polypeptide comprises mutations at amino acid positions 243, 264, 267 and 328
of the Fc
region, wherein the numbering is according to the EU index as in Kabat. In one
embodiment,
the mutations are F243AN264A/S267E/L328F. In one embodiment, the Fe-containing
polypeptide is an antibody fragment comprising SEQ ID NO:7. In another
embodiment, the Fe-
containing polypeptide is an antibody fragment comprising SEQ ID NO:8. In
another embodiment,
the Fc-containing polypeptide of the invention is an antibody fragment
comprising SEQ ID NO:17.
In another embodiment, the Fe-containing polypeptide is an antibody fragment
consisting (or
consisting essentially of) SEQ ID NO:7 or SEQ ID NO:8 or SEQ ID NO:17. In one
embodiment, at
least 47 mole % of the N-glycans have the structure
NANA2Ga12G1cNAc2Man3G1cNAc2.
In one embodiment, the sialic acid residues in the sialylated N-glycans are
attached via an a-
2,6 linkage. In one embodiment, the sialic acid residues in the sialylated N-
glycans are
attached via an a-2,6 linkage and there is no detectable level of an a-2,3
linked sialic acid. In
one embodiment, the sialylated N-glycans will comprise no N-glycolylneuraminic
acid
(NGNA).
As utilized herein, the term "pharmaceutically acceptable" means a non-toxic
material that does not interfere with the effectiveness of the biological
activity of the active
ingredient(s), approved by a regulatory agency of the Federal or a state
government or listed
in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use
in animals
and, more particularly, in humans. The term "carrier" refers to a diluent,
adjuvant, excipient,
or vehicle with which the therapeutic is administered and includes, but is not
limited to such
sterile liquids as water and oils. The characteristics of the carrier will
depend on the route of
administration.
Pharmaceutical Formulations of therapeutic and diagnostic agents may be
prepared by mixing with acceptable carriers, excipients, or stabilizers in the
form of, e.g.,
lyophilized powders, slurries, aqueous solutions or suspensions (see, e.g.,
Hardman et al.
(2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics, McGraw-
Hill,
New York, NY; Gennaro (2000) Remington: The Science and Practice of Pharmacy,
Lippincott, Williams, and Wilkins, New York, NY; Avis, et al. (eds.) (1993)
Pharmaceutical
- 46 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Dosage Forms: Parenteral Medications, Marcel Dekker, NY; Lieberman, et al.
(eds.) (1990)
Pharmaceutical Dosage Forms: Tablets, Marcel Dekker, NY; Lieberman, etal.
(eds.) (1990)
Pharmaceutical Dosage Forms: Disperse Systems, Marcel Dekker, NY; Weiner and
Kotkoskie (2000) Excipient Toxicity and Safety, Marcel Dekker, Inc., New York,
NY).
The mode of administration can vary. Suitable routes of administration
include oral, rectal, transmucosal, intestinal, parenteral; intramuscular,
subcutaneous,
intradermal, intramedullary, intrathecal, direct intraventricular,
intravenous, intraperitoneal,
intranasal, intraocular, inhalation, insufflation, topical, cutaneous,
transdermal, or intra-
arterial.
In certain embodiments, the Fc-containing polypeptides of the invention can
be administered by an invasive route such as by injection (see above). In some
embodiments
of the invention, the Fc-containing polypeptides of the invention, or
pharmaceutical
composition thereof, is administered intravenously, subcutaneously,
intramuscularly,
intraarterially, intra-articularly (e.g. in arthritis joints), intratumorally,
or by inhalation,
aerosol delivery. Administration by non-invasive routes (e.g., orally; for
example, in a pill,
capsule or tablet) is also within the scope of the present invention.
In certain embodiments, the the Fc-containing polypeptides of the invention
can be administered by an invasive route such as by injection (see above). In
some
embodiments of the invention, the Fc-containing polypeptides of the invention,
or
pharmaceutical composition thereof, is administered intravenously,
subcutaneously,
intramuscularly, intraarterially, intra-articularly (e.g. in arthritis
joints), intratumorally, or by
inhalation, aerosol delivery. Administration by non-invasive routes (e.g.,
orally; for example,
in a pill, capsule or tablet) is also within the scope of the present
invention.
Compositions can be administered with medical devices known in the art. For
example, a pharmaceutical composition of the invention can be administered by
injection
with a hypodermic needle, including, e.g., a prefilled syringe or
autoinjector.
The pharmaceutical compositions of the invention may also be administered
with a needleless hypodermic injection device; such as the devices disclosed
in U.S. Patent
Nos. 6,620,135; 6,096,002; 5,399,163; 5,383,851; 5,312,335; 5,064,413;
4,941,880;
4,790,824 or 4,596,556.
The pharmaceutical compositions of the invention may also be administered
by infusion. Examples of well-known implants and modules form administering
pharmaceutical compositions include: U.S. Patent No. 4,487,603, which
discloses an
implantable micro-infusion pump for dispensing medication at a controlled
rate; U.S. Patent
- 47 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
No. 4,447,233, which discloses a medication infusion pump for delivering
medication at a
precise infusion rate; U.S. Patent No. 4,447,224, which discloses a variable
flow implantable
infusion apparatus for continuous drug delivery; U.S. Patent. No. 4,439,196,
which discloses
an osmotic drug delivery system having multi-chamber compartments. Many other
such
implants, delivery systems, and modules are well known to those skilled in the
art.
Alternately, one may administer the antibody in a local rather than systemic
manner, for example, via injection of the antibody directly into an arthritic
joint, often in a
depot or sustained release formulation. Furthermore, one may administer the
antibody in a
targeted drug delivery system, for example, in a liposome coated with a tissue-
specific
antibody, targeting, for example, arthritic joint or pathogen-induced lesion
characterized by
immunopathology. The liposomes will be targeted to and taken up selectively by
the afflicted
tissue.
The administration regimen depends on several factors, including the serum or
tissue turnover rate of the therapeutic antibody, the level of symptoms, the
immunogenicity of
the therapeutic antibody, and the accessibility of the target cells in the
biological matrix.
Preferably, the administration regimen delivers sufficient therapeutic
antibody to effect
improvement in the target disease state, while simultaneously minimizing
undesired side
effects. Accordingly, the amount of biologic delivered depends in part on the
particular
therapeutic antibody and the severity of the condition being treated. Guidance
in selecting
appropriate doses of therapeutic antibodies is available (see, e.g.,
Wawrzynczak (1996)
Antibody Therapy, Bios Scientific Pub. Ltd, Oxfordshire, UK; Kresina (ed.)
(1991)
Monoclonal Antibodies, Cytokines and Arthritis, Marcel Dekker, New York, NY;
Bach (ed.)
(1993) Monoclonal Antibodies and Peptide Therapy in Autoimmune Diseases,
Marcel
Dekker, New York, NY; Baert, et al. (2003) New Engl. J. Med. 348:601-608;
Milgrom etal.
(1999) New Engl. J. Med. 341:1966-1973; Slamon et al. (2001) New Engl. J. Med.
344:783-
792; Beniaminovitz etal. (2000) New Engl. J. Med. 342:613-619; Ghosh etal.
(2003) New
Engl. J. Med. 348:24-32; Lipsky etal. (2000) New Engl. J. Med. 343:1594-1602).
Determination of the appropriate dose is made by the clinician, e.g., using
parameters or factors known or suspected in the art to affect treatment.
Generally, the dose
begins with an amount somewhat less than the optimum dose and it is increased
by small
increments thereafter until the desired or optimum effect is achieved relative
to any negative
side effects. Important diagnostic measures include those of symptoms of,
e.g., the
inflammation or level of inflammatory cytokines produced. Preferably, a
biologic that will be
used is derived from the same species as the animal targeted for treatment,
thereby
-48-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
minimizing any immune response to the reagent. In the case of human subjects,
for example,
chimeric, humanized and fully human Fc-containing polypeptides are preferred.
Fc-containing polypeptides can be provided by continuous infusion, or by
doses administered, e.g., daily, 1-7 times per week, weekly, bi-weekly,
monthly, bimonthly,
quarterly, semiannually, annually etc. Doses may be provided, e.g.,
intravenously,
subcutaneously, topically, orally, nasally, rectally, intramuscular,
intracerebrally,
intraspinally, or by inhalation. A total weekly dose is generally at least
0.05 ug/kg body
weight, more generally at least 0.2 jig/kg, 0.5 jig/kg, 1 ug/kg, 10 ug/kg, 100
[tg/kg, 0.25
mg/kg, 1.0 mg/kg, 2.0 mg/kg, 5.0 mg/ml, 10 mg/kg, 25 mg/kg, 50 mg/kg or more
(see, e.g.,
Yang et al., New Engl. J. Med. 349:427-434 (2003); Herold et al., New Engl. J.
Med.
346:1692-1698 (2002); Liu et al., J. Neurol. Neurosurg. Psych. 67:451-456
(1999); Portielji et
al., Cancer Immunol. Immunother. 52:133-144 (2003). In other embodiments, an
Fc-
containing polypeptide of the present invention is administered subcutaneously
or
intravenously, on a weekly, biweekly, "every 4 weeks," monthly, bimonthly, or
quarterly
basis at 10, 20, 50, 80, 100, 200, 500, 1000 or 2500 mg/kg subject.
As used herein, the terms "therapeutically effective amount", "therapeutically
effective dose" and "effective amount" refer to an amount of an Fc-containing
polypeptide of
the invention that, when administered alone or in combination with an
additional therapeutic
agent to a cell, tissue, or subject, is effective to cause a measurable
improvement in one or
more symptoms of a disease or condition or the progression of such disease or
condition. A
therapeutically effective dose further refers to that amount of the Fc-
containing polypeptide
sufficient to result in at least partial amelioration of symptoms, e.g.,
treatment, healing,
prevention or amelioration of the relevant medical condition, or an increase
in rate of
treatment, healing, prevention or amelioration of such conditions. When
applied to an
individual active ingredient administered alone, a therapeutically effective
dose refers to that
ingredient alone. When applied to a combination, a therapeutically effective
dose refers to
combined amounts of the active ingredients that result in the therapeutic
effect, whether
administered in combination, serially or simultaneously. An effective amount
of a therapeutic
will result in an improvement of a diagnostic measure or parameter by at least
10%; usually
by at least 20%; preferably at least about 30%; more preferably at least 40%,
and most
preferably by at least 50%. An effective amount can also result in an
improvement in a
subjective measure in cases where subjective measures are used to assess
disease severity.
- 49 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
EXAMPLE 1
Strains and Reagents
Escherichia coli strains TOP10 or DH5a (Invitrogen, CA) were used for
recombinant DNA work. Restriction endonucleases, DNA modification enzymes and
PNGase F were obtained from New England Biolabs, Ipswich, MA. Oligonucleotides
were
ordered from Integrated DNA Technologies, Coralville, IA.
EXAMPLE 2
Construction of IgG1 Fc muteins and Pichia pastoris recombinant expression
vector
The preparation of double and quadruple Fc muteins of the 1F11 IgG1
monoclonal antibody in Pichia pastoris was carried out using the sequences and
protocols
listed below.
A. Heavy and light chains
The heavy and light chain sequences, SEQ ID NOS: 1 and 2, respectively,
used for the preparation of the wildtype (parent) 1F11 monoclonal IgG1
antibody are as set
forth below. The heavy chain sequence used for the preparation of the 1F11
double mutein
antibody is set forth in SEQ ID NO:3. The heavy chain sequence used for the
preparation of
the 1F11 quadruple mutein antibody is shown in SEQ ID NO:4. All light chain
sequence for
all antibodies was the same. The heavy and light chains were codon optimized
according to
Pichia pastoris codon usage and synthesized by GenScript USA Inc. 860
Centennial Ave.
Piscataway, NJ 08854.
B. Signal sequence
The signal sequence of an a-Mating Factor predomain was fused in frame to
the 5' end of the light or heavy chain by PCR fusion. The sequence was codon
optimized as
described above. A Kozak sequence AAACG was added to the 5' end of the
methionine and
an EcoRl site was added before the Kozak sequence for cloning purposes. The
DNA
sequence (SEQ ID NO: 5) and amino acid (SEQ ID NO: 6) translation are as shown
below.
C. Recombinant plasmids for expression IgG1 and IgG1 Fc muteins
The heavy and light chains with the fused signal sequence of IgG1 and its
muteins were cloned under Pichia pastoris A0X1 promoter and in front of S.
cerevisiae Cyc
terminator, respectively. The expression cassette of the completed heavy and
light chains was
put together into the final expression vector. Genomic insertion into Pichia
pastoris was
achieved by linearization of the vector with Spel and targeted integration
into the Trp2 site.
-50-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
A summary of the plasmids used herein is given below in Table 1. A graphic
representation of the final expression plasmid for the 1F11 quadruple Fc
mutein is set forth in
Figure 1.
Table 1
Plasmid Description
pGLY9535 1F11 in wild type IgG1 expression plasmid
pGLY9543 1F11 in IgG1 F243AN264A double mutein expression
plasmid
1F11 in IgG1 F243A/V264A/S267E/L328F quadruple mutein
pGLY8068 expression plasmid
EXAMPLE 3
Glycoengineered Pichia GFI5.0 and GFI6.0 hosts for producing 1F11 and its Fc
muteins
Two different glycoengineered Pichia hosts were applied in this invention,
GFI5.0 and GFI 6Ø Following the procedures disclosed in Gerngross, US
7,029,872 and
Gerngross, US 7,449,308, one can construct vectors that are useful for
genetically engineering
lower eukaryotic host cells such that they are capable of expressing a desired
polypeptide
having a desired N-glycoform as the predominant species. GFI 5.0 and GFI6.0
strains were
engineered from NRRL11430 (American Type Culture Collection (ATCC), P.O. Box
1549,
Manassas, VA 20108, USA) according to the methods described in Hamilton et
al., Science,
313: 1441-1443 (2006) and Hamilton US 2006/0286637. The engineered Pichia
pastoris
strain GFI5.0 is capable of producing proteins with a biantennary N-glycan
structure with
terminal galactose. The genotype of the GFI5.0 strain used herein, YGLY17108,
is as
follows: ura5 A::ScSUC2 och 1 A: :lacZ bmt2A:lacZIK1M7VN2-2
mnn4L1A::lacZIMmSLC35A3
pno14::lacZ ADEL :lacZ/FB8/NA10/MmSLC35A3 his 1 : : lacZ-URA 5-
lacZ/XB33/SpGALE/DmUGT argl ::HIS1/KD53/TC54PRO1::ARG1/A0X1-
ScMFpreTrMNS1URA6-LmSTT3d. The genotype of the engineered Pichia pastoris
strain
GFI 6.0, YGLY17159, is as follows: ura5A::ScSUC2 ochl A::lacZ
bmt2A:lacZIK1MNN2-2
mnn4L1A::lacZIMmSLC35A3 pno 1 A
mnn4A::1acZADELlacZ/NA10/MmSLC35A3/FB8his 1 A::lacZ/ScGAL 10/XB33/DmUGTargl A
::HISI/KD53/TC54bmt4A::lacZ bmt 1 A: :lacZ
bmt3A::1acZTRP2:ARG1/MmCST/HsGNE/HsCSS/HsSPS/MmST6-33stel3A::lacZITHIDS1
dap2A::NatRTRP5:HygRMmCST/HsGNE/HsCSS/HsSPS/MmST6-33 Vps 10-1A: :
A0X1p LmSTT3d pGAPDH-mPomGnT1-56. YGLY17159 is capable of producing proteins
with a biantennary N-glycan structure on which terminal a 2,6-linked sialic
acid is attached to
galactose.
-51 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
The abbreviations used to describe the genotypes are commonly known and
understood by those skilled in the art, and include the following
abbreviations:
ScSUC2 S. cerevisiae Invertase
OCH1 Alpha-1,6-mannosyltransferase
K1MNN2-2 K. lactis UDP-GleNAc transporter
BMT1 Beta-mannose-transfer (beta-mannose elimination)
BMT2 Beta-mannose-transfer (beta-mannose elimination)
BMT3 Beta-mannose-transfer (beta-mannose elimination)
BMT4 Beta-mannose-transfer (beta-mannose elimination)
MNN4L1 MNN4-like 1 (charge elimination)
MmSLC35A3 Mouse homologue of UDP-GleNAc transporter
PNO1 Phosphomannosylation of N-glycans (charge elimination)
MNN4 Mannosyltransferase (charge elimination)
ScGAL10 UDP-glucose 4-epimerase
XB33 Truncated HsGalT1 fused to ScKRE2 leader
DmUGT UDP-Galactose transporter
KD53 Truncated DmMNSII fused to ScMNN2 leader
TC54 Truncated RnGNTII fused to ScMNN2 leader
NA10 Truncated HsGNTI fused to PpSEC12 leader
FB8 Truncated MmMNS1A fused to ScSEC12 leader
TrMDS1 Secreted T reseei MNS1
ADE1 N-succiny1-5-aminoimidazole-4-carboxamide ribotide
(SAICAR)
synthetase
MmCST Mouse CMP-sialic acid transporter
HsGNE Human UDP-GleNAc 2-epimerase/N-acetylmannosamine kinase
HsCSS Human CMP-sialic acid synthase
HsSPS Human N-acetylneuraminate-9-phosphate synthase
MmST6-33 Truncated Mouse alpha-2,6-sailyltransferase fused to
ScKRE2 leader
LmSTT3d Catalytic subunit of oligosaccharyltransferase from
Leishmania major
EXAMPLE 4
Yeast transformation and screening
The glycoengineered GFI5.0 and GS6.0 strains were grown in YPD rich media
(yeast extract 1%, peptone 2%and 2%dextrose), harvested in the logarithmic
phase by
- 52 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
centrifugation, and washed three times with ice-cold 1 M sorbitol. One to five
tig of a Spel
digested plasmid was mixed with competent yeast cells and electroporated using
a Bio-Rad
Gene Pulser XcellTM (Bio-Rad, 2000 Alfred Nobel Drive, Hercules, CA 94547)
preset Pichia
pastoris electroporation program. After one hour in recovery rich media at 24
C, the cells
were plated on a minimal dextrose media (1.34% YNB, 0.0004% biotin, 2%
dextrose, 1.5%
agar) plate containing 300 g/m1 Zeocin and incubated at 24 C until the
transformants
appeared.
To screen for high titer strains, 96 transformants were inoculated in buffered
glycerol-complex medium (BMGY) and grown for 72 hours followed by a 24 hour
induction
in buffered methanol-complex medium (BMMY). Secretion of antibody was assessed
by a
Protein A beads assay as follows. Fifty micro liter supernatant from 96 well
plate cultures
was diluted 1:1 with 50 mM Tris pH 8.5 in a non-binding 96 well assay plate.
For each 96
well plate, 2 ml of magnetic BioMag Protein A suspension beads (Qiagen,
Valencia, CA)
were placed in a tube held in a magnetic rack. After 2-3 minutes when the
beads collected to
the side of the tube, the buffer was decanted off. The beads were washed three
times with a
volume of wash buffer equal to the original volume (100 mM Tris, 150 mM NaC1,
pH 7.0)
and resuspended in the same wash buffer. Twenty p1 of beads were added to each
well of the
assay plate containing diluted samples. The plate was covered, vortexed gently
and then
incubated at room temperature for 1 hour, while vortexing every 15 minutes.
Following
incubation, the sample plate was placed on a magnetic plate inducing the beads
to collect to
one side of each well. On the Biomek NX Liquid Handler (Beckman Coulter,
Fullerton, CA),
the supernatant from the plate was removed to a waste container. The sample
plate was then
removed from the magnet and the beads were washed with 100 I wash buffer. The
plate was
again placed on the magnet before the wash buffer was removed by aspiration.
Twenty
loading buffer (Invitrogen E-PAGE gel loading buffer containing 25 mM NEM
(Pierce,
Rockford, IL)) was added to each well and the plate was vortexed briefly.
Following
centrifugation at 500 rpm on the Beckman Allegra 6 centrifuge, the samples
were incubated
at 99 C for five minutes and then run on an E-PAGE high-throughput pre-cast
gel
(Invitrogen, Carlsbad, CA). Gels were covered with gel staining solution (0.5
g Coomassie
G250 Brilliant Blue, 40% Me0H, 7.5% Acetic Acid), heated in a microwave for 35
seconds,
and then incubated at room temperature for 30 minutes. The gels were de-
stained in distilled
water overnight. High titer colonies were selected for further Sixfors
fermentation screening
- 53 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
described in detail in Example 5. A summary of the IgG1 wild type and Fc
mutein producing
strains is given below in Table 2.
Table 2
Strain Description
YGLY21351 GFI5.0 strain producing 1F11 IgG1 in GFI 5.0
YGLY23258 GFI6.0 strain producing 1F11 IgG1 F243AN264A
YGLY25265 GFI6.0 strain producing 1F11 IgG1F243A/V264A/S267E/L328F
YGLY25266
YGLY25267
YGLY25268 .
YGLY25269
EXAMPLE 5
Production of antibody in shake flasks
The strains were cultivated in 500m1 shake flasks with 300m1 of 2% BMGY
media and shaked at 24oC for 3 days.
Protocol for induction of shake flasks: Collect the total volume of each
culture
(300m1) into falcon tubes and spin at 2500 rpm for 5 minutes. Pour away
supernatant and
resuspend cell pellets in a final volume of 150m12% BMMY and 360u1PMTi4 (stock
concentration 0.65mg/m1). Transfer to a fresh 500m1 shake flask and shake at
24oC for 2
days. Spin down the induced cultures and collect the supernatant into fresh
falcon tubes.
The secreted antibodies were purified by protein A column using GE
Healthcare, STREAMLINE rProtein A (catalog no. 17-1281-01) and BioRad poly-
prep
chromatography columns (10m1) (catalog no. 731-1550). The following buffers
were used:
= Wash buffer #1: 20mM Tris pH 7.0, 1M NaC1
= Wash buffer #2: 20mM Tris pH 7.0
= Neutralization buffer: 1M Tris pH 8.0- pH 9.0
= Elution buffer: 100 mM or 50 mM Sodium Citrate pH 3.0
= Cleaning solution: 6M Urea in water.
The purification protocol is as follows:
= Add 500u1 of STREAMLINE rProtein A beads to each BioRad column. The beads
should be in 20% ethanol. The composition of the bead slurry should be 50%
beads,
50% liquid.
= Once the protein A beads are in the column they should be washed with
5mls of Wash
buffer #2 (discard the flow through)
- 54 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
= Add 10mls of supernatant to the BioRad column. During this step the
antibodies will
bind to the protein A beads. (discard the flow through)
= Wash away undesired excess proteins by adding 5mls of Wash buffer #1 to
the
column. (discard the flow through)
= Wash the column again by adding 5mls of Wash buffer #2 (discard the flow
through)
= Add lml of Neutralization buffer to the 15m1 protein collection tube.
= Place the BioRad column into the 15ml collection tube.
= Add 3mls of Elution buffer to the BioRad column. This will remove the
desired
antibodies from the protein A beads.
= Collect the eluted protein in the 15m1 protein collection tube.
= Determine the concentration of the eluted protein by Bradford assay (use
lOul of
protein for Bradford assay).
EXAMPLE 6
N-linked glycan analysis by HPLC
To quantify the relative amount of each glycoform, the N-glycosidase F
released glycans were labeled with 2-aminobenzidine (2-AB) and analyzed by
HPLC as
described in Choi et al., Proc. Natl. Acad. Sci. USA 100: 5022-5027 (2003) and
Hamilton et
al., Science 313: 1441-1443 (2006). Table 2 shows glycan profile of double
mutein and
quadruple mutein expression in GFI 6.0 host and 1F11 IgG1 in GFI 5.0 host.
These strains
were cultivated in shake flask. The results are shown in Table 2.
Table 2. Glycan profile of 1F11 IgGl, IgG1 double mutein and IgG1 quadruple
mutein
Strains M4 GO M5 G1 M6 G2 Al AlH A2 Sum
YGLY21351 0.3 12.9 24.1 39.3 23.3
100.0
YGLY23258 1.8 1.8 3.6 24.6 32.7
35.6 100.0
YGLY25265
YGLY25267
48.3 8.3 43.4 100.0
YGLY25268 5.6 5.9 39.4
41.4 100.0
YGLY25269 26.8 29.8
38.2 100.0
EXAMPLE 7
FcyR binding assay
- 55 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Fcy receptor binding assays were carried out as described (Shields, et al.
(2001) J. Biol. Chem. 276:6591-6604) with minor modifications. High protein
binding 96-
well plates (Corning Costar) were coated with 100uL per well of Fcy receptor
solutions in
PBS at the following concentrations: 1 g/mL for both FcyRI (R&D Systems) and
FcyRlIa
(Pichia pastoris produced), 2 g/mL for FcyRIIb (P. pastoris produced),
0.8gg/mL for
FcyRIIIa-F158, and 0.4ug/mL for FcyRIIIa-V158 (both P. pastoris produced). All
Pichia
pastoris produced receptors were expressed and purified as described (Li, et
al. (2006) Nature
Biotechnology 24:210-215). Monomeric antibody samples added to the FcyRI plate
were
serially diluted in assay diluent (lx PBS, 1% BSA, and 0.05% Tween20) and
100uL per well
was added to the plate. Antibody samples prepared for the remaining receptors
require a one
hour dimerization step with half molar ratio of goat anti-human IgG F(ab')2
F(ab')2 that is
conjugated with alkaline phosphatase for detection. These dimerized
F(ab')2/antibody
complexes were also serially diluted and 1004 per well were added to the
remaining
receptor plates and all plates were incubated for one hour at room
temperature. FcyRI bound
sample antibody was detected using the same goat anti-human IgG F(ab')2
alkaline
phosphatase conjugated F(ab')2. Sample antibody binding was quantified by
measuring
excitation at 340nm and emission at 465nm after 18 hour incubation with
SuperPhos, 4-MUP
Fluorescence AP Substrate Detection System (Virolabs).
The results are shown in Tables 3-7. Each table represents the result of an
individual experiment. A graphical representation of the data presented in
Table 3, is found
in Figure 3. A graphical representation of the data presented in Table 4, is
found in Figure 4.
A graphical representation of the data presented in Table 5, is found in
Figure 5. A graphical
representation of the data presented in Table 6, is found in Figure 6. A
graphical
representation of the data presented in Table 7, is found in Figure 7.
Table 3. IC50 COMPARISON TABLE
Samples FcyRIIB FcyRIIIA LF FcyRIIIA LV
Herceptin (CHO; dimer) 0.38 0.22 0.07
YGLY21351 (dimer) 0.12 0.02 0.04
YGLY25269 (dimer) 0.03 no binding no binding
YGLY25267 (dimer) 0.01 no binding no binding
YGLY23258 (dimer) no binding 0.60 0.09
Table 4. IC50 COMPARISON TABLE
Samples IFcyRIIB FeyRIIIA LF FeyRIIIA LV
- 56 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Herceptin (CHO; dimer) 0.54 0.21 0.09
YGLY21351 (dimer) -0.17 0.02 0.03
YGLY25266 (dimer) 0.02 no binding no binding
YGLY25268 (dimer) 0.02 no binding no binding
YGLY23258(dimer) no binding 0.69 0.12
Table 5. IC50 COMPARISON TABLE
Samples FcyRIIB FcyRIIIA LF FcyRIIIA LV
Herceptin (CHO; dimer) 0.43 0.24 0.09
YGLY21351 (dimer)) 0.18 0.02 0.04
YGLY25269 (dimmer) 0.03 no binding no binding
YGLY25267 (dimer) 0.01 no binding no binding
YGLY23258 (dimer) no binding 0.94 0.13
Table 6. IC50 COMPARISON TABLE
Samples FcyRIIB FcyRIIIA LF FcyRIIIA LV
Herceptin (CHO; dimer) 0.63 0.25 0.10
YGLY21351(dimer)) 0.20 0.02 0.03
YGLY25266 (dimer) 0.01 no binding no binding
YGLY25268 (dimer) 0.01 no binding no binding
YGLY23258 (dimer) no binding 1.17 0.16
Table 7. 1050 COMPARISON TABLE
Samples FcyRI FcyRIIA FcyRIIB FcyRIIIA LF FcyRIIIA LV
Herceptin (CHO; dimer) 0.09 0.04 0.72 0.23 0.08
YGLY21351 (dimer) 0.08 0.04 0.23 0.02 0.03
YGLY25269 (dimer) 0.50 no binding 0.03 no binding no binding
-YGLY25269 (monomer) 0.42 no binding 1.26 no binding no binding
YGLY23258 (dimer) 0.26 no binding no binding 1.31 0.16
Table 8 summarizes the relative decrease in binding to the various receptors
when compared to the wildtype parent antibody produced in GS 5Ø This data is
also
presented in Figure 8.
Table 8.
Samples FcyRI FcyRIIA
FcyRIIB FcyRIIIA LF FcyRIIIA LV
IgG1 G56.0 DM 4, 4 no binding no binding 1 8-60 1 2-6
IgG1 GS6.0 QM 1 5-6 no binding T 4-13
no binding no binding
1 indicates decreased affinity fold
I indicates increased affinity fold
- 57 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
EXAMPLE 8
Antigen affinity assay
The binding affinity of the anti-PCSK9 antibodies of the invention was
measured on a Biacore T100 instrument with a carboxymethylated dextran (CM5,
cat# BR-
1006-68) chip and lx HBS-EP+ (10mM HEPES, 150mM NaC1, 3mM EDTA, and 0.05%
Surfactant P20) as the running buffer. The CM5 chip was immobilized on all
flowcells with
mouse anti-human IgG (Fc specific) according to the Biacore Human Antibody
Capture Kit
(Cat# BR-1008-39) to -7000RU. Anti-PCSK9 antibodies were captured on the chip
to
-500RU followed by analyte injections of wild-type human PCSK9 from 64.1nM to
2.0nM.
Each flowcell was regenerated between each analyte injection with 3M MgC1 for
40 seconds
at lOuL/min. Data was analyzed with Biacore T100 Evaluation Software using the
1:1
binding model using at least 5 point concentration range sensogram.
As shown in Table 9, antigen affinity for the various anti-PCSK9 antibodies
made by the materials and methods hereinare similar.
Table 9.
ka (1/M*s) kd (Vs) KD (nM)
Samples Association Dissociation
Dissociation Average KD (nM)
Rate Rate Constant
1F11 1.52E+05 4.99E-04 3.29
2.80 0.70
1F11 1.77E+05 4.07E-04 2.30
YGLY25267-1 2.01E+05 6.34E-04 3.16
YGLY25267-2 1.93E+05 6.57E-04 3.41
YGLY25267-3 2.06E+05 6.61E-04 3.20
3.64 0.4681
YGLY25267-4 1.82E+05 7.79E-04 4.29
YGLY25267-5 1.94E+05 7.12E-04 3.68
YGLY25267-6 1.80E+05 7.37E-04 4.09
EXAMPLE 9
Construction, expression and characterization of additional IgG1 Fc muteins
Additional Fc muteins (Table 10) were constructed according to the methods
of Example 2.
- 58 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Table 10.
SEQ ID NO.
Expression (heavy
chain/
Strain Parent Plasmid Target light
chain)
YGLY29110 YGLY22834 pGLY8068 anti-PCSK9 1F11 QM 4/2
YGLY29111 YGLY22834 pGLY8068 anti-PCSK9 1F11 QM 4/2
YGLY29105 YGLY22834 pGLY11555 anti-IgE QM 11/10
YGLY29098 YGLY22834 pGLY11554 anti-IgE DM 9/10
YGLY30176 YGLY22834 pGLY11515 anti-TNF alpha QM 12/13
YGLY30177 YGLY22834 pGLY11515 anti-1NF alpha QM 12/13
YGLY30178 YGLY22834 pGLY11515 anti-TNF alpha QM 12/13
YGLY28722 YGLY22834 pGLY11539 human IgG1 Fe ("hFc") QM 8
YGLY28725 YGLY22834 pGLY11539 human IgG1 Fe ("hFc") QM 8
The expression vectors containing the Fe muteins described in Table 10 were
transformed in glycoengineered Pichia pastoris strain YGLY22834 (GFI6.0) which
is capable
adding alpha 2, 6 sialic acid onto bi-antennary galactoslyated glycan (G2).
The genotype of
the YGLY22834 strain used is as follows: ura5A::ScSUC2 ochlA::lacZ
bmt2A::lacZ/K1MNN2-2; mnn4L1A::lacZ/MmSLC35A3 pnolA mnn4A::lacZ;
ADE1::lacZ/NA10/MmSLC35A3/FB8; hislA::lacZ/ScGAL10/XB33/DmUGT;
argl A::HIS1/KD53/TC54; bmt4A::lacZ bmtlA::lacZ bmt3A::lacZ;
TRP2::ARG1/MmCST/HsGNE/HsCSS/HsSPS/MmST6-33; stel3A::lacZ-URA5-
lacZ/TrMDS1 dap2A::NatR; TRP5::HygRMmCST/HsGNE/HsCSS/HsSPS/MmST6-33;
vps10-1::pA0X1-LmSTT3d.
Fe mutein antibodies were produced and purified as indicated in Examples 4-
5.
The glycosylation of the Fe muteins produced herein was quantified by HPLC
as described in Example 6, and the results are shown in Table 11.
Table 11. Glycan profile of the Fe muteins described in Table 10.
Glycans
Strains
mAbs G2 Al All
A2 Others Sum
1F11 QM YGLY29110 1 10 3 84 2
100
1F11 QM YGLY29111 1 13 5 75 5
100
Anti-IgE QM YGLY29105 18 10 60 12
100
Anti-IgEr DM YGLY29098 6 30 5 54 4
100
Anti-TNF QM YGLY30176 1 9 4 83 2
100
Anti-TNF QM YGLY30177 2 14 7 60 15
100
Anti-TNF QM YGLY30178 3 21 5 67 3
100
hFc QM YGLY28722 2 19 8 66 2
100
- 59 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
hFc QM YGLY28725 5 15 3 63 14 100
The affinity of these Fc muteins towards the various FcyRs was measured as
described in Example 7, and the results are shown in Tables 12 and 13.
The samples identified as "Anti-TNF IgG1 GS5" refers to an antibody
The samples identified as "1F11 IgG1 G55.0" refers to an antibody comprising
the heavy chain of SEQ ID NO:1, and the light hcain of SEQ ID NO:2, produced
in a
recombinant Pichia pastoris strain GFI 5Ø
The sample identified as "Anti-TNF DM" refers to an antibody comprising the
heavy chain of SEQ ID NO:16 and the light chain of SEQ ID NO:13, produced in a
recombinant Pichia pastoris strain YGLY22834 (GFI6.0).
The sample identified as "1F11 DM" refers to an antibody comprising the
heavy chain of SEQ ID NO:3 and the light chain of SEQ ID NO:2, produced in a
recombinant
The sample identified as "human IgG Fe DM" or "hFC DM" refers to a
polypeptide comprising the amino acid sequence of SEQ ID NO:14 produced in a
recombinant Pichia pastoris strain YGLY22834 (GFI6.0) according to the methods
described
above.
Table 12.
Affinity (nM)
Samples (nM) (nM) (nM)
(nM)
STDEV STDEV STDEV
STDEV
Anti-TNF IgG1 GS5.0 463 16 53 7 1053 6
683 52
Anti-TNF DM 3265 + 440 195 9
5558 416 1552 212
Anti-TNF QM 8067 2984 5188 1563 5927 380 33 1 8
1F11 IgG1 GS5.0 203 14 0.15 0.02
892 26 545 40
1F11 DM 2517 122 97 6
11293 2531 1693 264
1F11 QM 20967 12115 5722 1723 8147 4875 29 5
hFc DM 3473 68
107 11 10040 628 2793 300
hFc QM 30583 813 6403 203 9133 556 28 2
n = 3
*Note - any value reported greater than ¨5000nM should be intrepreted as a
poor binder rather
than relying on the actual value because these values are significantly
outside the testing
concentration range.
Table 13.
- 60 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Fold Changes in Affinity
FcgRIIIA LF FcgRIIIA LV FcgRIIA H FcgRIIB
Samples (nM) (nM) (nM) (nM)
STDEV STDEV STDEV STDEV
Anti-TNF IgG1 GS5.0 1 1 1 1
Anti-TNF DM 71 41 51 21
Anti-TNF QM 171 981 6J, 211
1F11 IgG1 GS5.0 1 1 1 1
1F11 DM 121 6471 131 31
1F11 QM 1031 >100001 91 191
hFc DM 1 1 1 1
hFc QM 91 591 1 1001
i - indicates improved binding affinity
1 - indicates decreased binding affinity
EXAMPLE 10
Effect of the Fe muteins in a model of immune thrombocytopenia ("ITP")
ANIMALS: Fourteen (14) week old C57BL/6 female mice were obtained from
Taconic Farms.
MODEL INDUCTION: The mice are dose with the reagesnts listed in Table
14 by intravenous (iv) infusion on day 0. After 24 hours, ITP is induced with
a 2 ps iv dose
of anti-CD41 antibody (MWReg30) obtained from BD Biosystems. Twentyfour (24)
hours
after ITP induction, platelet counts are done on whole blood samples using a
Hemavet blood
cell analyzer.
Table 14.
Group/ Reagent Strain Dose (mpk)
hFc QM sialylated YGLY28725 50 mpk
hFc QM asialylated YGLY28725 50 mpk
hFc DM sialylated YGLY30175 50 mpk
hFc DM asialylated YGLY27893 50 mpk
GAMMAGARD N/A 1000 mpk
GAMMUNEX N/A 1000 mpk
All Groups n=4
Materials
The sample identified as ""hFC DM sialylated" refers to a polypeptide
comprising the amino acid sequence of SEQ ID NO:14 produced in a recombinant
Pichia
pastoris strain YGLY22834 (GFI6.0)
- 61 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
The sample identified as ""hFC DM asialylated" refers to a polypeptide
comprising the amino acid sequence of SEQ ID NO:14 produced in a recombinant
Pichia
pastoris strain YGLY22834 (GFI6.0) , which was subsequently treated in vitro
with
neuramindase to produce an asialylated form of the protein with terminal
galactose.
The sample identified as ""hFC QM sialylated" refers to a polypeptide
comprising the amino acid sequence of SEQ ID NO:8 produced in a recombinant
Pichia
pastoris strain YGLY22834 (GFI6.0).
The sample identified as ""hFC QM asialylated" refers to a polypeptide
comprising the amino acid sequence of SEQ ID NO:8 produced in a recombinant
Pichia
pastoris strain YGLY22834 (GFI6.0) , which was subsequently treated in vitro
with
neuramindase to produce an asialylated form of the protein with terminal
galactose.
In all cases the cultivation of the Pichia pastoris strains were erformed in
15 L
glass bioreactors. Briefly, two 3 L baffled seed flasks containing 500 mL of
BSGY medium
(4% glycerol, 1% yeast extract, 2% soytone, 100 mM potassium phosphate buffer,
pH 6.5,
100 mM D-sorbitol, 1.34% yeast nitrogen base, and 4x10-5 % biotin) were
inoculated with
yeast patches growing on agar plates. The flasks were incubated at 24 C and
180 rpm for
48 h to ensure exponential growth when the cells were transferred to the
bioreactor containing
BSGY medium, at a 10% volumetric ratio. Temperature was controlled at 24 C,
pH was
controlled at 6.5 with 28% ammonium hydroxide, and dissolved oxygen (DO) was
maintained at 20% of saturation at atmospheric pressure and 24 C by fixing
the airflow rate
at 0.7 vvm and cascading agitation. Depletion of the initial 40 g L-1 glycerol
was detected by
a rapid decrease in the oxygen uptake rate (OUR in mmol L-1 ICI) and was
followed by an
exponential 50% glycerol feed, starting at 5.3 g L-111-1 and increasing
exponentially at a rate
of 0.08 h-1. After 8h of glycerol fed-batch phase, the methanol induction in
oxygen-limited
environment was initiated. The DO cascade was turned off, and agitation was
set to a setpoint
of 460 rpm to achieve OUR of 20-25 mmol/L/h. After DO decreased to less than
1%, the first
1% (w/v) bolus shot of 100% methanol was delivered. All subsequent methanol 1%
(w/v)
bolus shots were triggered by rapid increases in DO indicating methanol
depletion.
The samples ( "hFC DM sialylated", "hFC DM asialylated", "hFC QM
sialylated" and "hFC QM sialylated") were purified using MabSelect (from GE
Healthcare
Life Sciences).
The N-glycan analysis of the "hFC DM sialylated" and "hFC QM sialylated"
samples was determined by HPLC, and had the following N-glycan
characteristics:
- 62 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
Glyeans
mAbs Al AlH A2
hFC DM sialylated 88 6 3
hFC QM sialylated 88 6 3
GAMMAGARD was obtained from Baxter Healthcare.
GAMMUNEX was obtained from by Talecris Biotherapeutics Inc.
The sample "ITP control" was anti-CD41 antibody (MWReg30) obtained from
BD Biosystems.
Results
The platelets value obtained are listed in Table 15 (K/ !IL) and plotted in
Figure 9. hFc DM sialylated and hFc QM sialylated showed statistically
significant
protection from ITP by One-way Anova analysis.
Naïve ITP hFc DM hFc DM hFc QM hFc QM
Cont Cont GAMMAGARD GAMMUNEXAsialylated Sialylated Asialylated Sialylated
1051. 208. 857. 713. 109. 795. 169. 545.
1078. 155. 796. 897. 182. 646. 89. 578.
1043. 130. 799. 811. 168. 827. 139. 622.
1086. 88. 813. 680. 142. 799. 145. 577.
- 63 -

CA 02834589 2013-10-28
W02012/162277
PCT/US2012/038915
SEQ DESCRIPTION SEQUENCE
ID
NO
I Heavy chain QVQLVQSGAEVKKPGASVKVSCKVSGYTFTDYYMNWVRQAPGQGLEWIG
(1F11 wt) DINPNNGGAIYNQKFKGRATLTVDKSTSTAYMELRSLRSDDTAVYYCTS
GIITEIAEDFWGQGTLVTVSSASTKGPSV
FPLAPSSKSTSGGTAAL
GCLVKDYFPEPVTVSWN
SGALTSGVHTFPAVLQS
SGLYSLSSVVTVPSSSL
GTQTYICNVNHKPSNTK
/DKKVEPKSCDKTHTCP
PCPAPELLGGPSVFLFP
PKPKDTLMISRTPEVTC
/VVDVSHEDPEVKFNWY
/DGVEVHNAKTKPREEQ
YNSTYRVVSVLTVLHQD
WLNGKEYKCKVSNKALP
APIEKTISKAKGQPREP
QVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVE
WESNGQPENNYKTTPPV
LDSDGSFFLYSKLTVDK
SRWQQGNVFSCSVMHEA
LHNHYTQKSLSLSPG
2 Light chain DIQMTQSPSSLSASVGDRVTITCKASQNVGTNVVWYQQKPGKAPKALIH
(1F11) SASYRYSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYKTYPYTF
GQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQ
WKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEV
THQGLSSPVTKSFNRGEC
3 Heavy chain QVQLVQSGAEVKKPGASVKVSCKVSGYTFTDYYMNWVRQAPGQGLEWIG
(1F11 DINPNNGGAIYNQKFKGRATLTVDKSTSTAYMELRSLRSDDTAVYYCTS
double GIITEIAEDFWGQGTLVTVSSASTKGPSV
mutein) FPLAPSSKSTSGGTAAL
GCLVKDYFPEPVTVSWN
SGALTSGVHTFPAVLQS
SGLYSLSSVVTVPSSSL
GTQTYICNVNHKPSNTK
/DKKVEPKSCDKTHTCP
PCPAPELLGGPSVFLAP
PKPKDTLMISRTPEVTC
/VADVSHEDPEVKFNWY
/DGVEVHNAKTKPREEQ
YNSTYRVVSVLTVLHQD
WLNGKEYKCKVSNKALP
APIEKTISKAKGQPREP
QVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVE
WESNGQPENNYKTTPPV
LDSDGSFFLYSKLTVDK
SRWQQGNVFSCSVMHEA
LHNHYTQKSLSLSPG
4 Heavy chain QVQLVQSGAEVKKPGASVKVSCKVSGYTFTDYYMNWVRQAPGQGLEWIG
(1F11 DINPNNGGAIYNQKFKGRATLTVDKSTSTAYMELRSLRSDDTAVYYCTS
quadruple GIITEIAEDFWGQGTLVTVSSASTKGPSV
- 64 -

CA 02834589 2013-10-28
W02012/162277
PCT/US2012/038915
mutein) FPLAPSSKSTSGGTAAL
GCLVKDYFPEPVTVSWN
SGALTSGVHTFPAVLQS
SGLYSLSSVVTVPSSSL
GTQTYICNVNHKPSNTK
/DKKVEPKSCDKTHTCP
PCPAPELLGGPSVFLAP
PKPKDTLMISRTPEVTC
/VADVEHEDPEVKFNWY
/DGVEVHNAKTKPREEQ
YNSTYRVVSVLTVLHQD
WLNGKEYKCKVSNKAFP
APIEKTISKAKGQPREP
QVYTLPPSRDELTKNQV
SLTCLVKGFYPSDIAVE
WESNGQPENNYKTTPPV
LDSDGSFFLYSKLTVDK
SRWQQGNVFSCSVMHEA
LHNHYTQKSLSLSPG
DNA GAATTCGAAACGATGAGATTTCCTTCAATTTTTACTGCTGTTTTATTCG
sequence of CAGCATCCTCCGCATTAGCT
signal
sequence of
an a-Mating
Factor
predomain
6 Amino acid MRFPSIFTAVLFAASSALA
sequence of
signal
sequence of
an a-Mating
Factor
predomain
7 Fe domain TCPPCPAPELLGGPSVF
QM LAPPKPKDTLMISRTPE
/TCVVADVEHEDPEVKF
NWYVDGVEVHNAKTKPR
EEQYNSTYRVVSVLTVL
HQDWLNGKEYKCKVSNK
AFPAPIEKTISKAKGQP
REPQVYTLPPSRDELTK
NQVSLTCLVKGFYPSDI
AVEWESNGQPENNYKTT
PPVLDSDGSFFLYSKLT
/DKSRWQQGNVFSCSVM
HEALHNHYTQKSLSLSP
G K
8 Fe domain AEPKSCDKTHTCPPCPA
QM PELLGGPSVFLAPPKPK
DTLMISRTPEVTCVVAD
/EHEDPEVKFNWYVDGV
EVHNAKTKPREEQYNST
YRVVSVLTVLHQDWLNG
KEYKCKVSNKAFPAPIE
KTISKAKGQPREPQVYT
LPPSRDELTKNQVSLTC
LVKGFYPSDIAVEWESN
- 65 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
GQPENNYK T T P P V LDSD
GSF FLYSKL TVDK SR WQ
QGNVF S CS VMHEALHNH
YTQKSLSLSPGK
9 Anti-IgEDMEVQL V ES GGGL VQPGGS
heavychainLRL S C AVS GY S I T S GYS
WNWIR QAPGK GLE WV AS
I T YDGS TNY ADS V K GRF
T ISRDDSKNTF YLQMNS
= RAEED T AV Y YCAR GSHY
F GHWHF AV WGQG T L V TV
SS AS TKGPS VF PLAPS S
K S TSGGT A ALGCL VKDY
F PEP V T VS WNS GAL T SG
/HTF P AVLQSSGL YSLS
S V V TV PSS SL GTQ T Y IC
NVNHK PSNTKVDKKVEP
KSCDK THT CP P
PAPPEL
LGGPS VFL AP PK PKDTL
MISR T PEV T CV V ADVSH
EDPEVKFNWYVDGVEVH
NAK TK PREEQYNS T YR V
/S VL T VLHQDWLNGKEY
K CK V SNK ALP AP IEK T I
SKAKGQPREPQVY TL PP
SR DEL TKNQVSL T CLVK
GF YP SDI AVEWESNGQP
ENNYK T TPPVLDSDGSF
F L YS K L T VDK SR WQQGN
/F S CS VMHEALHNHYTQ
KS LSL S PG
Anti- IgE DIQL T QS P
S S LS AS VGD
light chainR V T I T CR AS QS VD YDGD
S YMNWYQQK PGK APKLL
I YAAS YLES GVP SR F SG
S GS G TDF TL T I S S LQPE
DF AT Y YCQQSHEDPY TF
GQGTK V E IKR TV A AP S V
F IF PP SDEQLK SG T AS V
/ CL LNNF YPRE AK VQWK
/DNALQSGNSQES V TEQ
DS KDS T YSL SS TL TLSK
= DYEEK HK V YACEV THQG
L S S P VT K S FNR GE C
11 Anti-IgEQMEVQL V ES GGGL VQPGGS
heavy chainLR L S C AVS GY S I T S GYS
WNWIR QAPGK GLE WV AS
I T YDGS TNY ADS V K GR F
TISRDDSKNTF YLQMNS
= RAEEDT AV Y YCAR GSHY
F GHWHF AV WGQG T L V TV
S S AS TKGPS V F PLAPS S
KS TSGGTAALGCLVKDY
F PEP V TV S WNS GAL T SG
/HTF P AVLQSSGL YSLS
S V V TV P S S SLGTQT Y I C
NVNHK PSNTKVDKKVEP
KSCDK TH T CP PCP APEL
- 66 -

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
LGGPS VFLAP PK PKD T L
MISR TPEV T CV V ADVEH
EDPEVKFNWYVDGVEVH
NAK TK PR EEQYNS T YR V
/S VL T VLHQDWLNGKEY
K CK V SNK AF P AP I EK T I
SK AK GQPREPQV Y TLPP
SR DEL TKNQVSL T CLVK
GF YP SDI AVEWESNGQP
ENNYK T TPPVLDSDGSF
F L YSKL TVDK SR WQQGN
/F S CS VMHEALHNHYTQ
KSLSL S PG
12 Anti-TNFQMEVQL V ES GGGL VQPGR S
heavychainLRLSCAASGF TF DD Y AM
HWVRQAPGKGLEWVS Al
T WNS GHIDY ADS V EGR F
T I SRDNAKNS L YLQMNS
= RAEED T AV Y YCAK VS YL
S T AS S LDYWGQGT L V TV
S S AS TKGPS VF PLAPS S
K S T SGGT A ALGCL VKD Y
F PEPV TVS WNS GAL TSG
/HTFPAVLQSSGL YSLS
SVV TV P S S SLGTQ T Y IC
NVNHK PSNTK VDKK V E P
KSCDK TH T CP PCP APEL
LGGPS VF LAPPKPKDTL
MISR T PEV T CV V ADVEH
EDPEVKFNWYVDGVEVH
NAK TK PR EEQYNS T YR V
/S VL T VLHQDWLNGKEY
KCKVSNK AF P APIEK T I
SK AK GQPREPQV Y T L PP
SR DEL TKNQVSL T CLVK
GF YP SDI AV EWESNGQP
ENNYK T TPPVLDSDGSF
F L YSKL TVDK SR WQQGN
/F S CS VMHEALHNHY TQ
K SLSL S PG
13 Anti-TNFQMDIQM T QS P 5 S L S AS V GD
1 ight chain
R V T I T CR AS QGIRNYL A
WYQQK PGK APK LL I Y AA
S TLQS GVP SR F S GS GS G
TDF TL T IS S VAT A T
YYCQR YNR AP YTF GQGT
KVEIKR TV AAP S VF IF P
P SDEQLK SG T AS V V CLL
NNF Y PR EAK VQWK VDNA
LQS GNSQES V TEQDSKD
S T YSL SS TL TLSK ADYE
KHK V Y ACEV THQGLSSP
- 67 -

CA 02834589 2013-10-28
WO 2012/162277 PCT/US2012/038915
/ TK SF NR GEC
14 Fc domain AEPKSCDK TH T CP PCP A
DM PELLGGPS VF L AP PK PK
DTLMISR T PEV T CVV AD
/SHEDPEVKFNWYVDGV
EVHNAK TKPREEQYNS T
YR V VS VL TVLHQDWLN
GKEYKCKVSNK AL PAPI
EK TISK AKGQPREPQVY
TLPPSRDEL TKNQVSL T
CLVK GE YPSDIAVEWES
NGQPENNYK T TPPVLDS
DGSFFL YSKL T VDK SR W
QQGNVFSCSVMHEALHN
HY TQKSLSLSPGK
15 Anti- TNF EVQLVESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGL
heavy
EWVSAITWNSGHIDYADSVEGRFTISRDNAKNSLY
chain (wt)
LQMNSLRAEDTAVYYCAKVSYLSTASSLDYWGQGTLVTVSSASTK
GPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN
VNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLG
GPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG
KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKN
QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPP
VLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSL
SLSPG
16 Anti-TNF EVQLVESGGGLVQPGRS
DM heavy LRLSCAASGFTFDDYAM
chain HWVRQAPGKGLEWVSAI
TWNSGHIDYADSVEGRF
TISRDNAKNSLYLQMNS
LRAEDTAVYYCAKVSYL
STASSLDYWGQGTLVTV
SSASTKGPSVFPLAPSS
KSTSGGTAALGCLVKDY
FPEPVTVSWNSGALTSG
/HTFPAVLQSSGLYSLS
SVVTVPSSSLGTQTYIC
NVNHKPSNTKVDKKVEP
KSCDKTHTCPPCPAPEL
LGGPSVFLAPPKPKDTL
MISRTPEVTCVVADVSH
EDPEVKFNWYVDGVEVH
NAKTKPREEQYNSTYRV
/SVLTVLHQDWLNGKEY
KCKVSNKALPAPIEKTI
SKAKGQPREPQVYTLPP
-68-

CA 02834589 2013-10-28
WO 2012/162277
PCT/US2012/038915
SR DEL TKNQVSL TCLVK
GF YP S DI AV EWESNGQP
ENNYK T T PP VLDS DGS F
FLYSK L TVDK SR WQQGN
/ F S CS VMHE AL HNH Y TQ
KS LSL S PG
17 Fc domain E
PK SCDK THT CP P CP AP
QM EL
LGGPS V F L AP PK PKD
TLMISR T P EV T CV V ADV
EHEDPEVKFNWYVDGVE
/HNAK TK PR
STY T Y
R V VS V L TVLHQDWLNGK
EYKCK VSNKAF PAP IEK
T I SK AK GQPR EPQV Y TL
PPSRDEL TKNQVSL TCL
/KGF Y P SDI
EWES SNG
QPENNYK T T P P V LDSDG
SF F L YSKL T VDK SR WQQ
GNVF S CS VMHEALHNHY
TQKSLSLSPGK
While the present invention is described herein with reference to illustrated
embodiments, it should be understood that the invention is not limited hereto.
Those having
ordinary skill in the art and access to the teachings herein will recognize
additional
modifications and embodiments within the scope thereof.
- 69 -

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Application Not Reinstated by Deadline 2017-05-24
Time Limit for Reversal Expired 2017-05-24
Inactive: Abandon-RFE+Late fee unpaid-Correspondence sent 2017-05-23
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2016-05-24
Inactive: Cover page published 2013-12-13
Inactive: IPC assigned 2013-12-09
Inactive: IPC assigned 2013-12-09
Inactive: Notice - National entry - No RFE 2013-12-05
Letter Sent 2013-12-05
Inactive: IPC assigned 2013-12-05
Application Received - PCT 2013-12-05
Inactive: First IPC assigned 2013-12-05
Inactive: IPC assigned 2013-12-05
Inactive: IPC assigned 2013-12-05
Inactive: IPC assigned 2013-12-05
Inactive: IPC assigned 2013-12-05
BSL Verified - No Defects 2013-10-29
BSL Verified - Defect(s) 2013-10-29
Inactive: Sequence listing - Amendment 2013-10-29
Amendment Received - Voluntary Amendment 2013-10-28
National Entry Requirements Determined Compliant 2013-10-28
Application Published (Open to Public Inspection) 2012-11-29

Abandonment History

Abandonment Date Reason Reinstatement Date
2016-05-24

Maintenance Fee

The last payment was received on 2015-04-15

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
MF (application, 2nd anniv.) - standard 02 2014-05-22 2013-10-28
Basic national fee - standard 2013-10-28
Registration of a document 2013-10-28
MF (application, 3rd anniv.) - standard 03 2015-05-22 2015-04-15
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MERCK SHARP & DOHME CORP.
Past Owners on Record
DONGXING ZHA
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2013-10-27 69 4,072
Drawings 2013-10-27 9 427
Claims 2013-10-27 4 133
Abstract 2013-10-27 1 62
Representative drawing 2013-10-27 1 21
Description 2013-10-28 69 4,072
Cover Page 2013-12-12 1 41
Notice of National Entry 2013-12-04 1 193
Courtesy - Certificate of registration (related document(s)) 2013-12-04 1 102
Courtesy - Abandonment Letter (Maintenance Fee) 2016-07-04 1 171
Reminder - Request for Examination 2017-01-23 1 118
Courtesy - Abandonment Letter (Request for Examination) 2017-07-03 1 164
PCT 2013-10-27 5 165

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :